[go: up one dir, main page]

WO2025205612A1 - Method for propagating regulatory cells - Google Patents

Method for propagating regulatory cells

Info

Publication number
WO2025205612A1
WO2025205612A1 PCT/JP2025/011469 JP2025011469W WO2025205612A1 WO 2025205612 A1 WO2025205612 A1 WO 2025205612A1 JP 2025011469 W JP2025011469 W JP 2025011469W WO 2025205612 A1 WO2025205612 A1 WO 2025205612A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
regulatory
agonist
cell population
tgf
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/JP2025/011469
Other languages
French (fr)
Japanese (ja)
Inventor
新 金子
翔一 入口
義明 葛西
淳志 松田
敬子 関谷
崇之 佐藤
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Takeda Pharmaceutical Co Ltd
Kyoto University NUC
Original Assignee
Takeda Pharmaceutical Co Ltd
Kyoto University NUC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Takeda Pharmaceutical Co Ltd, Kyoto University NUC filed Critical Takeda Pharmaceutical Co Ltd
Publication of WO2025205612A1 publication Critical patent/WO2025205612A1/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues

Definitions

  • the present invention relates to a method for producing a cell population in which primary regulatory T cells (Treg) have expanded, a cell population containing regulatory T cells obtained by the method, a pharmaceutical product containing the cell population containing regulatory T cells, and the like.
  • regulatory T cells Treg
  • GvHD graft-versus-host disease
  • Regulatory T cells are broadly classified into two types: naturally occurring regulatory T cells (nTreg, or thymic Treg: tTreg) and inducible regulatory T cells (iTreg).
  • nTregs are naturally generated in the thymus, while iTregs are known to be induced to differentiate from naive T cells in peripheral blood by antigen stimulation and cytokines such as IL-2 and TGF- ⁇ .
  • cytokines such as IL-2 and TGF- ⁇ .
  • Non-Patent Document 1 Non-Patent Document 1
  • TGF- ⁇ Non-Patent Documents 1, 3, 6, 8, 11
  • regulatory T cells can be expanded with high efficiency while maintaining their suppressive function.
  • [5] The method according to any one of [1] to [4], wherein the TNFR2 agonist is an anti-TNFR2 agonist antibody.
  • [6] The method according to [2], [4] or [5], wherein the TGF- ⁇ R agonist is TGF- ⁇ .
  • [7] The method according to any one of [1] to [6], wherein the culture in the step (1) is further carried out in the presence of at least one agonist selected from the group consisting of a CD30 agonist, a CD3 agonist, and a CD28 agonist.
  • [9b] The method according to any one of [1] to [9a], wherein the regulatory T cells are Helios + cells.
  • a method for expanding a cell population containing regulatory T cells comprising: (1A) A method comprising the step of culturing a cell population containing primary regulatory T cells in the presence of IL-2, IL-4, IL-33 and a TNFR2 agonist. [10a] The step (1A) The method according to [10], further performed in the presence of a TGF- ⁇ R agonist. [10b] The step (1A) The method according to [10], further performed in the presence of IL-3.
  • [10c] The step (1A) The method according to [10], further performed in the presence of IL-3 and a TGF- ⁇ R agonist.
  • [10d] The method according to any one of [10] to [10c], wherein the TNFR2 agonist is an anti-TNFR2 agonist antibody.
  • [10e] The method according to [10a], [10c] or [10d], wherein the TGF- ⁇ R agonist is TGF- ⁇ .
  • [10f] The method according to any one of [10] to [10e], wherein the culture in the step (1A) is further carried out in the presence of at least one agonist selected from the group consisting of a CD30 agonist, a CD3 agonist, and a CD28 agonist.
  • [10f1] The method according to any one of [10] to [10f], wherein in the culture of the step (1A), the culture is further performed in the presence of at least one agonist selected from the group consisting of a CD30 agonist, a CD3 agonist, and a CD28 agonist, and then in the absence of these agonists.
  • the regulatory T cells are CD25 + /FOXP3 + cells.
  • the CD25 + /FOXP3 + cells are CD4 + cells.
  • [10i] The method according to any one of [10] to [10f1], wherein the regulatory T cells are CD25 + /CD127 ⁇ cells.
  • [10j] The method according to any one of [10] to [10i], wherein the regulatory T cells are Helios + cells.
  • [10k] (2) The method according to any one of [1] to [10j], comprising a step of introducing a foreign gene (particularly a gene for expressing a chimeric antigen receptor) into a cell population containing primary regulatory T cells or a cell population containing regulatory T cells obtained by the method according to any one of [1] to [10j].
  • [11] A cell population containing regulatory T cells obtained by the method according to any one of [1] to [10k].
  • a pharmaceutical comprising a cell population containing regulatory T cells according to [11].
  • the pharmaceutical agent according to [12] which is used for the prevention and/or treatment of abnormally enhanced immune response.
  • a method for preventing and/or treating abnormally enhanced immune responses comprising administering a cell population containing the regulatory T cells described in [11] to a subject in need thereof.
  • FIG. 1 is a graph showing the change in cell number over time when primary cultured Tregs were expanded in a medium containing IL-2, IL-3, IL-4, IL-33, TGF- ⁇ 1, and anti-TNFR2 antibody.
  • Optimized Medium containing IL-2, IL-3, IL-4, IL-33, TGF- ⁇ 1, and anti-TNFR2 antibody.
  • Control Medium not containing IL-3, IL-4, IL-33, TGF- ⁇ 1, or anti-TNFR2 antibody.
  • This figure shows the results of examining the expression levels of proteins expressed in Tregs expanded in a medium containing IL-2, IL-3, IL-4, IL-33, TGF- ⁇ 1, and an anti-TNFR2 antibody. All dot plots in the figure represent the CD3 + CD4 + CD8 ⁇ population. The values on the graph indicate the proportion of Tregs (CD3 + CD4 + CD8 ⁇ CD25 + FOXP3 + ) in each cell population.
  • the left figure shows the results of flow cytometry analysis of a cell population expanded in a medium containing IL-2, IL-3, IL-4, IL-33, TGF- ⁇ 1, and an anti-TNFR2 antibody after co-culture with allogeneic T cells derived from human PBMCs, and the right graph shows the inhibition rate (%) of target cell division.
  • E:T indicates the ratio of Treg to human T cell numbers.
  • FIG. 10 is a graph showing the change in cell number over time when primary cultured Tregs were expanded in a medium containing only IL-2, or in a medium containing IL-2, IL-3, IL-4, IL-33, TGF- ⁇ 1, and an anti-TNFR2 antibody from which any one of IL-3, IL-4, IL-33, and TGF- ⁇ 1 had been removed.
  • This figure shows the results of examining the expression levels of proteins expressed in Tregs expanded in a medium containing IL-2, IL-3, IL-4, IL-33, TGF- ⁇ 1, and an anti-TNFR2 antibody, but omitting any one of IL-3, IL-4, IL-33, and TGF- ⁇ 1.
  • All dot plots in the figure represent the CD3 + CD4 + CD8 ⁇ population.
  • the values on the graph indicate the proportion of Tregs (CD3 + CD4 + CD8 ⁇ CD25 + FOXP3 + ) in each cell population.
  • 1 is a graph showing the rate (%) of inhibition of target cell division after co-culture with human PBMC-derived allogeneic T cells using a cell population expanded in a medium containing IL-2, IL-3, IL-4, IL-33, TGF- ⁇ 1, and an anti-TNFR2 antibody but from which any one of IL-3, IL-4, IL-33, and TGF- ⁇ 1 has been removed.
  • E:T indicates the ratio of the number of Tregs to the number of human T cells.
  • 1 is a graph showing the change in cell number over time when primary cultured Tregs were expanded in a medium containing IL-2, IL-3, IL-4, IL-33, TGF- ⁇ 1, and an anti-TNFR2 antibody, or in a medium containing the above medium composition minus IL-3, TGF- ⁇ 1, and anti-TNFR2 antibody.
  • culture refers to maintaining and/or growing cells in an in vitro environment.
  • “Culturing” refers to maintaining and/or growing cells outside a tissue or outside the body, for example, in a cell culture dish or flask.
  • culturing a cell population in the presence of a substance refers to culturing a cell population in a medium containing the substance.
  • examples of such culturing include culturing in a medium containing the substance alone, or in which the substance coexists with other differentiation-inducing factors.
  • adding the substance to the medium it can be added directly to the medium, or the substance can be dissolved in an appropriate solvent just before use and then added to the medium.
  • the substance can also be immobilized on the surface of a substrate or carrier during culturing and then cultured.
  • negative (-) means that the expression level of a protein or gene is below the lower limit of detection by any or all of the above-mentioned known techniques, or that the level of expression is low.
  • the lower limit of detection for protein or gene expression may vary depending on the technique.
  • the level of protein or gene expression (whether low or high) can be determined by comparing it with the results of control cells measured under the same conditions. For example, the level of CD25 expression in a certain cell population can be determined using flow cytometry, by comparing the CD25 expression level in that cell population with the CD25 expression level in PBMC-derived Tconv (control: known to be a low CD25 expressor). If expression equivalent to that of the control is observed, it is determined to be low expression, and if expression is higher than that of the control cells, it is determined to be high expression.
  • the term "marker” refers to a protein or its gene that is specifically expressed on the cell surface, in the cytoplasm, in the nucleus, etc., of a specific cell type.
  • the marker is preferably a "cell surface marker.”
  • a “cell surface marker” refers to a protein expressed on the cell surface that can be labeled (stained) with a fluorescent substance, making it easy to detect, concentrate, isolate, etc., cells expressing the cell surface marker.
  • the cell surface marker refers to a gene that is specifically expressed (positive marker) or not expressed (negative marker) in a specific cell type; specifically, a substance that is produced (positive marker) or not produced (negative marker) as mRNA resulting from transcription of the gene in the genome, or as a protein resulting from translation of that mRNA.
  • Such cell surface markers can be detected using immunological assays using antibodies specific to the cell surface markers, such as ELISA, immunostaining, and flow cytometry.
  • expression is defined as the transcription and/or translation of a specific nucleotide sequence driven by a promoter within a cell.
  • regulatory T cells refers to T cells that, upon stimulation via a T cell receptor, have the ability to inhibit the activation of effector T cells and are responsible for suppressing immune responses (immune tolerance). Regulatory T cells are usually CD25 + /FOXP3 + cells or CD25 + /CD127 - cells, among which CD4 + or CD8 + cells are present.
  • regulatory T cells may be CD4 + cells (i.e., CD4 + /CD25 + /FOXP3 + or CD4 + /CD25 + /CD127 - ) or CD8 + cells (i.e., CD8 + /CD25 + /FOXP3 + or CD8 + /CD25 + /CD127 - ), with CD4 + cells being more preferred.
  • the transcription factor FOXP3 is known as a master regulator of CD4 + /CD25 + regulatory T cells.
  • Regulatory T cells may also be positive for Helios, CTLA4 (Cytotoxic T Lymphocyte (associated) Antigen 4) (CD152), CD39, and CD73, which are known to be indicators of the suppressive function of regulatory T cells.
  • regulatory T cells expanded and “expansion of regulatory T cells” refer to an increase in the number (absolute number) of regulatory T cells in a cell population compared to before culture or a control such as cells obtained without carrying out the present invention, and mean, for example, an increase of at least 1.1-fold, 1.2-fold, 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 300-fold, 500-fold, 1000-fold, 2000-fold, 3000-fold, 4000-fold, 5000-fold, 6000-fold, 7000-fold, 8000-fold, 9000-fold, 10000-fold, 15000-fold, 20000-fold, 50000-fold, or 100000-fold compared to before culture or the control.
  • the number (absolute number) of regulatory T cells in a cell population produced by the present invention is increased by at least 1.1-fold, 1.2-fold, 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 300-fold, 500-fold, 1000-fold, 2000-fold, 3000-fold, 4000-fold, 5000-fold, 6000-fold, 7000-fold, 8000-fold, 9000-fold, 10000-fold, 15000-fold, 20000-fold, 50000-fold, or 100000-fold compared to the number of cells before culture or in a control.
  • Nucleotide analogs may be naturally occurring or unnatural molecules, such as sugar-modified nucleotide analogs (e.g., nucleotide analogs substituted with 2'-O-methylribose, nucleotide analogs substituted with 2'-O-propylribose, nucleotide analogs substituted with 2'-methoxyethoxyribose, nucleotide analogs substituted with 2'-O-methoxyethylribose, nucleotide analogs substituted with 2'-O-[2-(guanidinium)ethyl]ribose, nucleotide analogs substituted with 2'-fluororibose, and bridged artificial nucleotides.
  • sugar-modified nucleotide analogs e.g., nucleotide analogs substituted with 2'-O-methylribose, nucleotide analogs substituted with 2'-O-propylribo
  • a nucleic acid derivative can be any molecule in which another chemical substance has been added to the nucleic acid in order to improve nuclease resistance, stabilization, affinity with complementary nucleic acid strands, cell permeability, or visualization, compared to nucleic acids.
  • Specific examples include 5'-polyamine-added derivatives, cholesterol-added derivatives, steroid-added derivatives, bile acid-added derivatives, vitamin-added derivatives, Cy5-added derivatives, Cy3-added derivatives, 6-FAM-added derivatives, and biotin-added derivatives.
  • the method of the present invention for producing a cell population in which regulatory T cells have expanded (sometimes referred to herein simply as the "production method of the present invention") is characterized by comprising the following steps: (1) Culturing a cell population containing primary regulatory T cells in the presence of interleukin (IL)-2, IL-4, IL-33, and a TNFR2 agonist.
  • IL interleukin
  • Step (1) can also be carried out in the presence of a TGF- ⁇ R agonist and/or IL-3 in addition to IL-2, IL-4, IL-33, and a TNFR2 agonist. That is, step (1) can be carried out in the presence of IL-2, IL-3, IL-4, IL-33, and a TNFR2 agonist; in the presence of IL-2, IL-4, IL-33, a TGF- ⁇ R agonist, and a TNFR2 agonist; or in the presence of IL-2, IL-3, IL-4, IL-33, a TGF- ⁇ R agonist, and a TNFR2 agonist. From the perspective of regulatory T cell proliferation, step (1) is preferably carried out in the absence of an mTOR inhibitor (e.g., rapamycin).
  • an mTOR inhibitor e.g., rapamycin
  • IL-4, IL-3, IL-2, and IL-33 are preferably derived from mammals, and particularly preferably from humans.
  • IL-2, IL-3, IL-4, and IL-33 may be natural products isolated and purified from mammals (particularly humans), or may be artificially produced using genetic engineering techniques (which may include amino acid substitutions, deletions, insertions, and/or additions).
  • the TNFR2 agonist is preferably an anti-TNFR2 agonist antibody.
  • the antibody may be a functional fragment thereof, such as Fd, Fv, Fab, F(ab'), F(ab) 2 , F(ab') 2 , single-chain Fv (scFv), diabody, triabody, tetrabody, and minibody.
  • Antibodies include those derived from animals such as mouse, rat, cow, rabbit, goat, sheep, and guinea pig.
  • the antibody isotype is not particularly limited, and examples of isotypes include IgG (IgG1, IgG2, IgG3, IgG4), IgA, IgD, IgE, and IgM.
  • the antibody may be either a monoclonal antibody or a polyclonal antibody, preferably a monoclonal antibody, and may also be a humanized antibody, a chimeric antibody, a multispecific antibody (e.g., a bispecific antibody), etc.
  • Antibodies can be produced by known methods, for example, by constructing an expression vector containing a nucleic acid encoding the antibody, culturing a transformant into which the nucleic acid has been introduced, or culturing a hybridoma that produces the antibody.
  • IL-2, IL-3, IL-4, IL-33, TGF- ⁇ R agonists, and TNFR2 agonists can be used in their free state or in the form of a salt.
  • salts include salts with inorganic bases such as sodium salt, magnesium salt, potassium salt, calcium salt, and aluminum salt; salts with organic bases such as methylamine salt, ethylamine salt, and ethanolamine salt; salts with basic amino acids such as lysine, ornithine, and arginine; and ammonium salts.
  • the concentration of IL-2 in the culture medium is not particularly limited, and is, for example, 0.1 to 1000 ng/mL, preferably 10 to 500 ng/mL, and more preferably 50 to 200 ng/mL.
  • the concentration of IL-4 in the culture medium is not particularly limited, and is, for example, 0.01 to 100 ng/mL, preferably 10 to 100 ng/mL.
  • the concentration of IL-33 in the culture medium is not particularly limited, and is, for example, 0.1 to 100 ng/mL, preferably 10 to 100 ng/mL.
  • the concentration of the TNFR2 agonist in the medium is not particularly limited and can be adjusted appropriately depending on the type of TNFR2 agonist used.
  • the concentration of the TNFR2 agonist is, for example, 0.0001 to 100 ⁇ g/mL, preferably 0.01 to 10 ⁇ g/mL.
  • the concentration of IL-3 in the culture medium is not particularly limited, and is, for example, 0.01 to 100 ng/mL, preferably 10 to 100 ng/mL.
  • the medium used in the culture of step (1) is a basal medium that is used for culturing animal cells and contains the aforementioned IL-2, IL-4, IL-33, and TNFR2 agonist (as well as IL-3, TGF- ⁇ R agonist, etc.).
  • the basal medium is not particularly limited as long as it can be used for culturing animal cells, and examples include AIM V, X-VIVO-15, NeuroBasal, EGM2, TeSR, BME, BGJb, CMRL 1066, Glasgow MEM, Improved MEM Zinc Option, IMDM, 199 Medium, Eagle's MEM, ⁇ MEM, DMEM, Ham, RPMI-1640, and Fischer's Medium. Any one of these media may be used alone, or two or more may be used in combination.
  • the medium may contain serum or may be serum-free.
  • the medium may also contain serum substitutes (e.g., albumin, transferrin, Knockout Serum Replacement (KSR), fatty acids, insulin, collagen precursors, trace elements, 2-mercaptoethanol, 3'-thiolglycerol, ITS supplement, B27TM supplement, etc.).
  • serum substitutes e.g., albumin, transferrin, Knockout Serum Replacement (KSR), fatty acids, insulin, collagen precursors, trace elements, 2-mercaptoethanol, 3'-thiolglycerol, ITS supplement, B27TM supplement, etc.
  • KSR Knockout Serum Replacement
  • the medium may contain one or more substances such as lipids, amino acids (non-essential amino acids, etc.), L-glutamine, vitamins, growth factors, cytokines, anti-CD3 antibodies, anti-CD30 antibodies, anti-CD28 antibodies, antibiotics, antioxidants, pyruvic acid, buffers, and inorganic salts. It is preferable to use a chemically defined medium that does not contain unknown components such as serum, as this reduces differences between medium lots and allows for the preparation of cells of consistent quality.
  • substances such as lipids, amino acids (non-essential amino acids, etc.), L-glutamine, vitamins, growth factors, cytokines, anti-CD3 antibodies, anti-CD30 antibodies, anti-CD28 antibodies, antibiotics, antioxidants, pyruvic acid, buffers, and inorganic salts. It is preferable to use a chemically defined medium that does not contain unknown components such as serum, as this reduces differences between medium lots and allows for the preparation of cells of consistent quality.
  • the culture period is also not particularly limited and can be appropriately determined by those skilled in the art while monitoring the number of regulatory T cells, etc., and is, for example, 7 days or more, preferably 14 days or more, more preferably 21 days or more, and even more preferably 28 days or more.
  • the upper limit of the culture period is not particularly limited and, for example, 42 days or less, preferably 35 days or less, and more preferably 28 days or less.
  • passages may be performed as many times as necessary to obtain the desired amount of regulatory T cells, and medium addition and replacement may be performed.
  • the culture of the present invention can be performed using a known CO2 incubator.
  • the culture vessel is not particularly limited and can be appropriately selected from plates, dishes, petri dishes, flasks, bags, bottles, tanks (culture vessels), bioreactors, etc. A vessel to which an anti-CD3 antibody is bound can be used as the culture vessel.
  • culture may be performed in the presence of IL-2, IL-4, IL-33, and a TNFR2 agonist (IL-3 and a TGF- ⁇ R agonist may also be added), and in the presence of at least one selected from the group consisting of a CD30 agonist, a CD3 agonist, and a CD28 agonist.
  • culture may be initially initiated in the presence of IL-2, IL-4, IL-33, and a TNFR2 agonist (IL-3 and a TGF- ⁇ R agonist may also be added), and in the presence of at least one selected from the group consisting of a CD30 agonist, a CD3 agonist, and a CD28 agonist, and then culture may be performed in the absence of these antibodies.
  • Step (1) may be repeated (by subculture).
  • the culture may be performed by first initiating culture (stimulating the cells) in the presence of at least one selected from the group consisting of a CD30 agonist, a CD3 agonist, and a CD28 agonist, followed by repeated culture in the absence of these antibodies.
  • the culture period for this repetition is, for example, 7 to 28 days, preferably 14 to 21 days.
  • the cells may be cultured (stimulated) in the presence of at least one selected from the group consisting of a CD30 agonist, a CD3 agonist, and a CD28 agonist for the first 3 days or so, followed by culture in a medium that does not contain these agonists for 4 to 11 days, and this culture unit may be repeated.
  • the number of culture repetitions is not particularly limited and can be determined appropriately by a person skilled in the art while monitoring the number of immune cells of the present invention, for example, 1 or more, 2 or more, 3 or more, 4 or more, 5 or more, 6 or more, or 7 or more.
  • the upper limit of the number of culture repetitions is, for example, 20 times.
  • the CD30 agonist is not particularly limited as long as it is a molecule that can transmit a signal from CD30 into the cell by specifically binding to CD30.
  • Examples of CD30 agonists include anti-CD30 agonist antibodies or functional fragments thereof, and CD30 ligands or functional fragments thereof.
  • the concentration of the CD30 agonist in the culture medium is not particularly limited and is adjusted appropriately depending on the type of CD30 agonist used.
  • the concentration of the CD30 agonist is, for example, 10 to 1000 ng/mL, preferably 100 to 500 ng/mL.
  • the CD3 agonist is not particularly limited, as long as it is a molecule that can transmit a signal from CD3 into the cell by specifically binding to CD3.
  • Examples of CD3 agonists include anti-CD3 agonist antibodies or functional fragments thereof, and CD3 ligands or functional fragments thereof.
  • the concentration of the CD3 agonist in the culture medium is not particularly limited and is adjusted appropriately depending on the type of CD3 agonist used.
  • the concentration of the CD3 agonist is, for example, 10 to 10,000 ng/mL, preferably 100 to 10,000 ng/mL, and more preferably 1,000 to 5,000 ng/mL.
  • the CD28 agonist is not particularly limited, as long as it is a molecule that can transmit a signal from CD28 into the cell by specifically binding to CD28.
  • Examples of CD28 agonists include anti-CD28 agonist antibodies or functional fragments thereof, and CD28 ligands or functional fragments thereof.
  • the concentration of the CD28 agonist in the culture medium is not particularly limited and is adjusted appropriately depending on the type of CD28 agonist used.
  • the concentration of the CD28 agonist is, for example, 10 to 10,000 ng/mL, preferably 100 to 10,000 ng/mL, and more preferably 500 to 5,000 ng/mL.
  • the cell population containing regulatory T cells used in the production method of the present invention is primary cultured cells isolated from biological tissues such as bone marrow, umbilical cord blood, and blood.
  • Primary cultured cells refer to cells that have been isolated from organs, cells, and tissues separated from a living body and seeded until the first passage.
  • Primary cultured cells can be obtained from organs and tissues by known methods such as enzyme treatment, physical dispersion, and the explant method.
  • the proportion (cell number) of regulatory T cells contained in the cell population containing primary regulatory T cells is, for example, 80% or more, preferably 90% or more, more preferably 95% or more, with an upper limit of, for example, 100% or less.
  • Regulatory T cells can be isolated from cells separated from biological tissues by known methods such as flow cytometry or magnetic cell separation.
  • the cell population containing primary regulatory T cells used in the production method of the present invention may be derived from humans or from mammals other than humans (non-human mammals), preferably humans.
  • non-human mammals include mice, rats, hamsters, guinea pigs, rabbits, dogs, cats, pigs, cows, horses, sheep, and monkeys.
  • the production method of the present invention may further include a step of separating regulatory T cells in order to enrich the obtained regulatory T cells. Separation of regulatory T cells can be carried out by known methods such as flow cytometry or magnetic cell separation.
  • the manufacturing method of the present invention makes it possible to produce a cell population containing regulatory T cells in which regulatory T cells have proliferated. In other words, the manufacturing method of the present invention makes it possible to expand and culture regulatory T cells.
  • Step (1A) can be carried out in the same manner as step (1).
  • the explanation regarding the production method of the present invention applies to the method for expanding the above-mentioned cell population containing regulatory T cells.
  • the regulatory T cells obtained by the production method and proliferation method of the present invention may be regulatory T cells into which an exogenous gene has been introduced.
  • exogenous gene is a gene introduced from outside to cause regulatory T cells to express a desired protein, and can be selected appropriately depending on the intended use of the regulatory T cells.
  • the foreign gene can be, for example, a gene for expressing a chimeric antigen receptor (CAR), which can further include genes for expressing cytokines and/or chemokines. Similar to common or known CARs, the CAR expressed by regulatory T cells is essentially composed of peptides from each of the following sites, linked, as necessary, via a spacer: (i) an antigen recognition site (e.g., a single-chain antibody) that recognizes a cell surface antigen on a cancer cell; (ii) a cell membrane-spanning domain; and (iii) a signal transduction domain that induces T cell activation.
  • the foreign gene can also be, for example, a gene for expressing an exogenous T cell receptor (TCR).
  • the means for introducing a foreign gene into cells is not particularly limited, and various known or common means can be used.
  • the foreign gene is introduced into cells using an expression vector and expressed.
  • the expression vector may be linear or circular, and may be a non-viral vector such as a plasmid, a viral vector, or a transposon-based vector.
  • the cells into which the foreign gene is introduced are not particularly limited, and may be at any stage. Examples include primary regulatory T cells before carrying out step (1) or (1A), and regulatory T cells obtained by the production method and expansion method of the present invention.
  • the expression vector can be introduced into cells by viral infection.
  • viral vectors include retroviral vectors, lentiviral vectors, adenoviral vectors, and adeno-associated viral vectors.
  • retroviral vectors include retroviral vectors, lentiviral vectors, adenoviral vectors, and adeno-associated viral vectors.
  • a corresponding commercially available kit can be used to transfect host cells with the vector containing the foreign gene and the packaging vector (plasmid) of each virus to produce a recombinant virus, and then the resulting recombinant virus can be used to infect cells.
  • a cell population containing regulatory T cells produced by the production method and expansion method of the present invention is useful for treating animals (particularly humans) with abnormally enhanced immune responses, including, but not limited to, X-linked immunodysregulation/polyendocrinopathy/enteropathy (IPEX) syndrome, graft-versus-host disease (GVHD), organ transplant rejection, autoimmune diseases, inflammatory diseases, and allergic diseases (hay fever, asthma, atopic dermatitis, eczema, food allergies, food hypersensitivity, urticaria, allergic rhinitis, allergic conjunctivitis, and drug allergies).
  • Regulatory T cells produced by the production method and expansion method of the present invention may be used for autologous or allogeneic transplantation. They may also be used in combination with other drugs.
  • the subject from whom the cells to be used in producing regulatory T cells are isolated has an HLA type that matches that of the subject to which the regulatory T cells will be administered, and it is even more preferable that the subject is the same as the subject to which the regulatory T cells will be administered.
  • a pharmaceutical comprising a cell population that includes regulatory T cells (hereinafter, sometimes referred to as the pharmaceutical of the present invention).
  • the pharmaceutical of the present invention is preferably produced as a parenteral formulation by mixing an effective amount of regulatory T cells with a pharmaceutically acceptable carrier according to known means (for example, the method described in the Japanese Pharmacopoeia).
  • the pharmaceutical of the present invention is preferably produced as a parenteral formulation such as an injection, suspension, or infusion.
  • Parenteral administration methods include intravenous, intraarterial, intramuscular, intraperitoneal, and subcutaneous administration.
  • Pharmaceutically acceptable carriers include, for example, solvents, bases, diluents, excipients, soothing agents, buffers, preservatives, stabilizers, suspending agents, isotonicity agents, surfactants, and solubilizing agents.
  • the dosage of the medicament of the present invention can be determined appropriately depending on various conditions, such as the patient's body weight, age, sex, and symptoms.
  • the number of cells administered per administration to a subject weighing 60 kg is typically 1 ⁇ 10 6 to 1 ⁇ 10 10 , preferably 1 ⁇ 10 7 to 1 ⁇ 10 9 , and more preferably 5 ⁇ 10 7 to 5 ⁇ 10 8.
  • the medicament may be administered once or multiple times.
  • the medicament of the present invention can be in a known form suitable for parenteral administration, such as an injection or infusion.
  • the medicament of the present invention may contain physiological saline, phosphate-buffered saline (PBS), a culture medium, or the like, to stably maintain the cells.
  • PBS phosphate-buffered saline
  • culture media examples include, but are not limited to, RPMI, AIM-V, and X-VIVO10.
  • the medicament may contain a pharmaceutically acceptable carrier (e.g., human serum albumin), a preservative, or the like, for stabilization purposes.
  • a pharmaceutically acceptable carrier e.g., human serum albumin
  • the medicament of the present invention is intended for use in mammals, including humans.
  • the medium contained, at final concentrations, FBS (15%, Corning), L-glutamine-penicillin-streptomycin solution (1/100, Invitrogen, Sigma-Aldrich), insulin-transferrin-selenium supplement (1/100, Invitrogen), ascorbic acid 2-phosphate (50 ⁇ g/mL, Sigma-Aldrich), IL-2 (100 ng/mL, PeproTech), and anti-TNFR2 antibody (3 ⁇ g/mL, Hycult
  • the ⁇ -MEM (Invitrogen) medium used contained IL-1 (60 ng/mL, BioLegend), IL-3 (60 ng/mL, BioLegend), IL-4 (30 ng/mL, BioLegend), IL-33 (30 ng/mL, BioLegend), and TGF- ⁇ 1 (5 ng/mL, BioLegend).
  • anti-CD28 antibody 1.5 ⁇ g/mL, BioLegend
  • anti-CD30 antibody 300 ng/mL, R&D Systems
  • a control group used medium containing the above medium composition but omitting anti-TNFR2 antibody, IL-3, IL-4, IL-33, and TGF- ⁇ 1.
  • the cells were cultured for 15 days, with the medium being changed on days 3, 6, 8, 11, and 13 after the start of culture, and the number of cells was counted and a line graph was created. The results are shown in Figure 2.
  • the cells were cultured for 4 days in ⁇ -MEM (Invitrogen) containing final concentrations of FBS (15%, Corning), L-glutamine-penicillin-streptomycin solution (1/100, Invitrogen, Sigma-Aldrich), insulin-transferrin-selenium supplement (1/100, Invitrogen), and ascorbic acid 2-phosphate (50 ⁇ g/mL, Sigma-Aldrich).
  • FBS FBS
  • L-glutamine-penicillin-streptomycin solution 1/100, Invitrogen, Sigma-Aldrich
  • insulin-transferrin-selenium supplement 1/100, Invitrogen
  • ascorbic acid 2-phosphate 50 ⁇ g/mL, Sigma-Aldrich
  • the primary cultured Tregs obtained in (1) were seeded at 1 x 10 cells/well onto a 96-well cell culture plate conjugated with anti-CD3 antibody (3 ⁇ g/mL, eBioscience) and cultured for 3 days at 37°C and 5.0% CO in a CO incubator. After further culture for 3 days, the cells were seeded onto a 48-well cell culture plate and continued to be cultured. After further culture for 3 days, the cells were seeded onto a 24-well G-Rex cell culture plate and continued to be cultured.
  • the medium contained, at final concentrations, FBS (15%, Corning), L-glutamine-penicillin-streptomycin solution (1/100, Invitrogen, Sigma-Aldrich), insulin-transferrin-selenium supplement (1/100, Invitrogen), ascorbic acid 2-phosphate (50 ⁇ g/mL, Sigma-Aldrich), IL-2 (100 ng/mL, PeproTech), and anti-TNFR2 antibody (3 ⁇ g/mL, Hycult).
  • the culture medium used was ⁇ -MEM (Invitrogen) containing IL-3 (60 ng/mL, BioLegend), IL-4 (30 ng/mL, BioLegend), IL-33 (30 ng/mL, BioLegend), and TGF- ⁇ 1 (5 ng/mL, BioLegend).
  • IL-3 60 ng/mL, BioLegend
  • IL-4 30 ng/mL, BioLegend
  • IL-33 (30 ng/mL, BioLegend
  • TGF- ⁇ 1 5 ng/mL, BioLegend
  • anti-CD28 antibody 1.5 ⁇ g/mL, BioLegend
  • anti-CD30 antibody 300 ng/mL, R&D Systems
  • a medium containing IL-2 but not IL-3, IL-4, IL-33, TGF- ⁇ 1, or anti-TNFR2 antibody (medium containing only IL-2) was used, or a medium obtained by removing any one of IL-3, IL-4, IL-33, and TGF- ⁇ 1 from the medium composition.
  • the culture was continued for 14 days, with medium changes on days 3, 6, 8, 11, and 13 after the start of culture, and the number of cells was counted and a line graph was created. The results are shown in FIG. 5.
  • the cell population obtained through expansion culture showed reduced proliferation ability on the 14th day of culture when IL-4 and IL-2 were removed from the above medium conditions.
  • the percentages of the CD25 + FOXP3 + population in each cell population were 62.8%, 50.5%, 65.8%, and 30.7%, respectively.
  • PBMCs derived from a donor other than Tregs and untreated or treated with anti-CD3 antibodies were stained with CellTrace Violet (Thermo Fisher Scientific) and used as target cells. These cells were co-cultured with Tregs expanded in a medium containing no cytokines (IL-3, IL-4, IL-33, or TGF- ⁇ 1) as described in (5).
  • cytokines IL-3, IL-4, IL-33, or TGF- ⁇ 1
  • the cells were cultured for 4 days in ⁇ -MEM (Invitrogen) containing final concentrations of FBS (15%, Corning), L-glutamine-penicillin-streptomycin solution (1/100, Invitrogen, Sigma-Aldrich), insulin-transferrin-selenium supplement (1/100, Invitrogen), and ascorbic acid 2-phosphate (50 ⁇ g/mL, Sigma-Aldrich).
  • FBS FBS
  • L-glutamine-penicillin-streptomycin solution 1/100, Invitrogen, Sigma-Aldrich
  • insulin-transferrin-selenium supplement 1/100, Invitrogen
  • ascorbic acid 2-phosphate 50 ⁇ g/mL, Sigma-Aldrich
  • the medium contained, at final concentrations, FBS (15%, Corning), L-glutamine-penicillin-streptomycin solution (1/100, Invitrogen, Sigma-Aldrich), insulin-transferrin-selenium supplement (1/100, Invitrogen), ascorbic acid 2-phosphate (50 ⁇ g/mL, Sigma-Aldrich), IL-2 (100 ng/mL, PeproTech), and anti-TNFR2 antibody (3 ⁇ g/mL, Hycult
  • the culture medium used was ⁇ -MEM (Invitrogen) containing IL-3 (60 ng/mL, BioLegend), IL-4 (30 ng/mL, BioLegend), IL-33 (30 ng/mL, BioLegend), and TGF- ⁇ 1 (5 ng/mL, BioLegend).
  • anti-CD28 antibody 1.5 ⁇ g/mL, BioLegend
  • anti-CD30 antibody 300 ng/mL, R&D Systems
  • a medium containing the above medium composition but omitting IL-3, TGF- ⁇ 1, and anti-TNFR2 antibody was also used.
  • the culture was continued for 22 days, with the medium being changed on days 3, 6, 8, 11, 13, 15, 18, and 20 after the start of culture, and the number of cells was counted and a line graph was created. The results are shown in Figure 8.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical & Material Sciences (AREA)
  • Cell Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biotechnology (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Wood Science & Technology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Transplantation (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Virology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Microbiology (AREA)
  • Hematology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

Disclosed are: a method for producing a cell population in which regulatory T cells have been propagated, the method comprising (1) a step for culturing a cell population containing primary regulatory T cells in the presence of interleukin (IL)-2, IL-4, IL-33, and a TNFR2 agonist; (1A) a method for propagating a cell population containing regulatory T cells, the method comprising a step for culturing a cell population containing primary regulatory T cells in the presence of IL-2, IL-4, IL-33, and a TNFR2 agonist; a cell population containing regulatory T cells obtained by the method; and a drug comprising the cell population containing regulatory T cells.

Description

制御性細胞の増殖方法Method for Proliferating Regulatory Cells

 本発明は、初代制御性T細胞(primary regulatory T cell(Treg))が増殖した細胞集団の製造方法、該方法により得られた制御性T細胞を含む細胞集団、当該制御性T細胞を含む細胞集団を含有してなる医薬等に関する。
(発明の背景)
The present invention relates to a method for producing a cell population in which primary regulatory T cells (Treg) have expanded, a cell population containing regulatory T cells obtained by the method, a pharmaceutical product containing the cell population containing regulatory T cells, and the like.
BACKGROUND OF THE INVENTION

 近年、制御性T細胞(regulatory T cell(Treg))は免疫応答抑制機能を持つため、様々な自己免疫疾患に対する治療目的として、制御性T細胞を用いた細胞治療(Treg therapy)の研究開発が世界で進められている。このように、制御性T細胞は、移植片対宿主病(GvHD)、自己免疫疾患の治療、炎症性疾患、アレルギー性疾患の治療及び予防に用いることが期待されている。 In recent years, research and development into cell therapy (Treg therapy) using regulatory T cells (Treg) has been progressing worldwide as a treatment for various autoimmune diseases, as regulatory T cells have the function of suppressing immune responses. In this way, regulatory T cells are expected to be used in the treatment and prevention of graft-versus-host disease (GvHD), autoimmune diseases, inflammatory diseases, and allergic diseases.

 制御性T細胞は、内在性制御性T細胞(naturally occurring regulatory T cell(natural Treg):nTreg、又はthymic Treg:tTreg)と誘導性制御性T細胞(inducible regulatory T cell(induced Treg):iTreg)の大きく2つに分類され、nTregは胸腺内で自然発生し、iTregは抗原刺激及びIL-2、TGF-βなどのサイトカインにより末梢血中のナイーブT細胞から分化誘導されることが知られている。これらの制御性T細胞は更に、その細胞に発現するマーカーの種類に従い細分化されている。 Regulatory T cells are broadly classified into two types: naturally occurring regulatory T cells (nTreg, or thymic Treg: tTreg) and inducible regulatory T cells (iTreg). nTregs are naturally generated in the thymus, while iTregs are known to be induced to differentiate from naive T cells in peripheral blood by antigen stimulation and cytokines such as IL-2 and TGF-β. These regulatory T cells are further subdivided according to the types of markers they express.

 制御性T細胞を生体より除去すると、各種の臓器特異的な自己免疫疾患が自然発症し、その際、制御性T細胞を移植すると、自己免疫疾患の発症が阻止されるため、制御性T細胞は、末梢での免疫自己寛容の維持に重要な働きをしていると考えられていた。その後、制御性T細胞は、自己免疫だけでなく、外来抗原による炎症、移植による拒絶反応、感染免疫、アレルギー、腫瘍免疫などのほとんどの免疫反応を抑制し得ることが明らかになってきた。また、現在、この制御性T細胞のマスターレギュレーターとして、転写因子FOXP3が明らかにされている。 When regulatory T cells are removed from the body, various organ-specific autoimmune diseases spontaneously develop. However, transplanting regulatory T cells in such cases prevents the onset of autoimmune diseases. Therefore, regulatory T cells were thought to play an important role in maintaining immune self-tolerance in the periphery. It has since become clear that regulatory T cells can suppress not only autoimmunity, but also most immune responses, including inflammation caused by foreign antigens, transplant rejection, infection immunity, allergies, and tumor immunity. Furthermore, the transcription factor FOXP3 has now been identified as the master regulator of regulatory T cells.

 FOXP3はTregのマスターレギュレーターであり、TregではFOXP3が恒常的に発現していること、及び抑制機能を有さない通常型T細胞(Tconv(conventional T細胞)、エフェクターT細胞、炎症性T細胞と称することもある)では恒常的な発現は見られないことが知られている。primary TregにおいてFOXP3の発現を維持したまま効率的に増殖させる方法に関する研究が行われ、それら発現維持及び細胞増殖の活性を有する化合物、サイトカイン、その組み合わせが報告されている。そのような化合物、サイトカインとしては、IL-2(非特許文献1-7)、IL-4(非特許文献2、8、9)、TNFR2アゴニスト抗体(非特許文献4、5、7、10)などがある。 FOXP3 is a master regulator of Tregs. It is known that FOXP3 is constitutively expressed in Tregs, but not in conventional T cells (also known as Tconv (conventional T cells), effector T cells, or inflammatory T cells) that do not have suppressive function. Research has been conducted on methods for efficiently expanding primary Tregs while maintaining FOXP3 expression, and compounds, cytokines, and combinations thereof that maintain expression and promote cell proliferation have been reported. Examples of such compounds and cytokines include IL-2 (Non-Patent Documents 1-7), IL-4 (Non-Patent Documents 2, 8, 9), and TNFR2 agonist antibodies (Non-Patent Documents 4, 5, 7, 10).

 さらに、そのような化合物、サイトカイン、及びその組み合わせとしては、IL-3(非特許文献1)、TGF-β(非特許文献1、3、6、8、11)などがある。 Further examples of such compounds, cytokines, and combinations thereof include IL-3 (Non-Patent Document 1) and TGF-β (Non-Patent Documents 1, 3, 6, 8, 11).

 Primary Tregに対する、IL-2、IL-4及びTNFR2アゴニストの効果についてはそれぞれ上記のように報告されているものの、IL-2、IL-4、IL-33及びTNFR2アゴニスト(さらにそこにIL-3又はTGF-βを加えた)の組み合わせが特に好ましいというような報告はない。 Although the effects of IL-2, IL-4, and TNFR2 agonists on primary Tregs have been reported as described above, there have been no reports suggesting that the combination of IL-2, IL-4, IL-33, and TNFR2 agonists (with the addition of IL-3 or TGF-β) is particularly preferable.

J Immunol(2011) 186:2262-2272J Immunol (2011) 186:2262-2272 Immunology.2009 Jul;127(3):338-344Immunology. 2009 Jul;127(3):338-344 PLoS One.,11(2):e0148474.(2016)PLoS One. , 11(2):e0148474. (2016) PLoS One.,11(5),e0156311(2016)PLoS One. , 11(5), e0156311 (2016) Front.Immunol.9:573(2018)Front. Immunol. 9:573 (2018) Clin.Exp.Immunol.,197(1):52-63.(2019)Clin. Exp. Immunol. , 197(1):52-63. (2019) Scientific Reports 3:3153(2013)Scientific Reports 3:3153 (2013) Immunology(2009) 128:e670-e678Immunology (2009) 128:e670-e678 Front.Immunol.8:1508.doi:10.3389/fimmu.2017.01508Front. Immunol. 8:1508. doi:10.3389/fimmu. 2017.01508 Sci.Signal.13,eaba9600(2020)Sci. Signal. 13, eaba9600 (2020) J Exp Med(2003) 198(12):1875-1886J Exp Med (2003) 198(12): 1875-1886

 本発明は、増殖させることが難しいことが知られている初代制御性T細胞を高い効率で、かつ、その抑制機能を維持したまま、増殖させることができる、制御性T細胞が増殖した細胞集団の製造方法を提供することを目的とする。 The present invention aims to provide a method for producing a cell population of expanded regulatory T cells, which can efficiently expand primary regulatory T cells, which are known to be difficult to expand, while maintaining their suppressive function.

 本発明者らは、上記目的を達成すべく鋭意研究を重ねた結果、IL-2、IL―4、IL-33及びTNFR2アゴニストの存在下で初代制御性T細胞を含む細胞集団を培養することで、高い効率、かつ、その抑制機能を維持させたまま、制御性T細胞を増殖させることができるという知見を得た。 As a result of extensive research conducted to achieve the above-mentioned objective, the inventors discovered that by culturing a cell population containing primary regulatory T cells in the presence of IL-2, IL-4, IL-33, and a TNFR2 agonist, regulatory T cells can be expanded with high efficiency while maintaining their suppressive function.

 本発明は、これら知見に基づき、更に検討を重ねて完成されたものであり、次の制御性T細胞が増殖した細胞集団の製造方法、制御性T細胞を含む細胞集団、医薬等を提供するものである。 The present invention was completed based on these findings and through further investigation, and provides the following methods for producing cell populations in which regulatory T cells have expanded, cell populations containing regulatory T cells, pharmaceuticals, etc.

[1]制御性T細胞が増殖した細胞集団の製造方法であって、
(1)インターロイキン(IL)-2、IL-4、IL-33及びTNFR2アゴニストの存在下で初代制御性T細胞を含む細胞集団を培養する工程を含む方法。
[2]前記工程(1)を、
更にTGF-βRアゴニストの存在下で行う、[1]に記載の方法。
[3]前記工程(1)を、
更にIL-3の存在下で行う、[1]に記載の方法。
[4]前記工程(1)を、
更にIL-3及びTGF-βRアゴニストの存在下で行う、[1]に記載の方法。
[5]前記TNFR2アゴニストが、抗TNFR2アゴニスト抗体である、[1]~[4]のいずれかに記載の方法。
[6]前記TGF-βRアゴニストが、TGF-βである、[2]、[4]又は[5]に記載の方法。
[7]前記工程(1)の培養において、更にCD30アゴニスト、CD3アゴニスト及びCD28アゴニストからなる群から選択される少なくとも1種の存在下で培養を行う、[1]~[6]のいずれかに記載の方法。
[7a]前記工程(1)の培養において、更にCD30アゴニスト、CD3アゴニスト及びCD28アゴニストからなる群から選択される少なくとも1種の存在下で培養を行った後、それらの非存在下で培養を行う、[1]~[6]のいずれかに記載の方法。
[8]前記制御性T細胞が、CD25/FOXP3細胞である、[1]~[7a]のいずれかに記載の方法。
[9]前記CD25/FOXP3細胞が、CD4である、[8]に記載の方法。
[9a]前記制御性T細胞が、CD25/CD127細胞である、[1]~[7a]のいずれかに記載の方法。
[9b]前記制御性T細胞が、Helios細胞である、[1]~[9a]のいずれかに記載の方法。
[10]制御性T細胞を含む細胞集団の増殖方法であって、
(1A)IL-2、IL-4、IL-33及びTNFR2アゴニストの存在下で初代制御性T細胞を含む細胞集団を培養する工程を含む方法。
[10a]前記工程(1A)を、
更にTGF-βRアゴニストの存在下で行う、[10]に記載の方法。
[10b]前記工程(1A)を、
更にIL-3の存在下で行う、[10]に記載の方法。
[10c]前記工程(1A)を、
更にIL-3及びTGF-βRアゴニストの存在下で行う、[10]に記載の方法。
[10d]前記TNFR2アゴニストが、抗TNFR2アゴニスト抗体である、[10]~[10c]のいずれかに記載の方法。
[10e]前記TGF-βRアゴニストが、TGF-βである、[10a]、[10c]又は[10d]に記載の方法。
[10f]前記工程(1A)の培養において、更にCD30アゴニスト、CD3アゴニスト及びCD28アゴニストからなる群から選択される少なくとも1種の存在下で培養を行う、[10]~[10e]のいずれかに記載の方法。
[10f1]前記工程(1A)の培養において、更にCD30アゴニスト、CD3アゴニスト及びCD28アゴニストからなる群から選択される少なくとも1種の存在下で培養を行った後、それらの非存在下で培養を行う、[10]~[10f]のいずれかに記載の方法。
[10g]前記制御性T細胞が、CD25/FOXP3細胞である、[10]~[10f1]のいずれかに記載の方法。
[10h]前記CD25/FOXP3細胞が、CD4である、[10g]に記載の方法。
[10i]前記制御性T細胞が、CD25/CD127細胞である、[10]~[10f1]のいずれかに記載の方法。
[10j]前記制御性T細胞が、Helios細胞である、[10]~[10i]のいずれかに記載の方法。
[10k](2)初代制御性T細胞を含む細胞集団、又は[1]~[10j]のいずれかに記載の方法により得られた、制御性T細胞を含む細胞集団に外来遺伝子(特にキメラ抗原受容体を発現するための遺伝子)を導入する工程を含む、[1]~[10j]のいずれかに記載の方法。
[11][1]~[10k]のいずれかに記載の方法により得られた、制御性T細胞を含む細胞集団。
[12][11]に記載の制御性T細胞を含む細胞集団を含有してなる、医薬。
[13]免疫反応の異常な亢進の予防及び/又は治療に用いるための、[12]に記載の医薬。
[14][11]に記載の御性T細胞を含む細胞集団を、それを必要とする対象に投与することを含む、免疫反応の異常な亢進の予防及び/又は治療方法。
[15]免疫反応の異常な亢進の予防及び/又は治療において使用するための、[11]に記載の御性T細胞を含む細胞集団。
[16]免疫反応の異常な亢進の予防及び/又は治療するための医薬の製造における、[11]に記載の御性T細胞を含む細胞集団の使用。
[1] A method for producing a cell population in which regulatory T cells have expanded, comprising:
(1) A method comprising the step of culturing a cell population containing primary regulatory T cells in the presence of interleukin (IL)-2, IL-4, IL-33 and a TNFR2 agonist.
[2] The step (1)
The method according to [1], further performed in the presence of a TGF-βR agonist.
[3] The step (1)
The method according to [1], further performed in the presence of IL-3.
[4] The step (1)
The method according to [1], further performed in the presence of IL-3 and a TGF-βR agonist.
[5] The method according to any one of [1] to [4], wherein the TNFR2 agonist is an anti-TNFR2 agonist antibody.
[6] The method according to [2], [4] or [5], wherein the TGF-βR agonist is TGF-β.
[7] The method according to any one of [1] to [6], wherein the culture in the step (1) is further carried out in the presence of at least one agonist selected from the group consisting of a CD30 agonist, a CD3 agonist, and a CD28 agonist.
[7a] The method according to any one of [1] to [6], wherein in the culture of the step (1), the culture is further performed in the presence of at least one agonist selected from the group consisting of a CD30 agonist, a CD3 agonist, and a CD28 agonist, and then in the absence of these agonists.
[8] The method according to any one of [1] to [7a], wherein the regulatory T cells are CD25 + /FOXP3 + cells.
[9] The method according to [8], wherein the CD25 + /FOXP3 + cells are CD4 + cells.
[9a] The method according to any one of [1] to [7a], wherein the regulatory T cells are CD25 + /CD127 cells.
[9b] The method according to any one of [1] to [9a], wherein the regulatory T cells are Helios + cells.
[10] A method for expanding a cell population containing regulatory T cells, comprising:
(1A) A method comprising the step of culturing a cell population containing primary regulatory T cells in the presence of IL-2, IL-4, IL-33 and a TNFR2 agonist.
[10a] The step (1A)
The method according to [10], further performed in the presence of a TGF-βR agonist.
[10b] The step (1A)
The method according to [10], further performed in the presence of IL-3.
[10c] The step (1A)
The method according to [10], further performed in the presence of IL-3 and a TGF-βR agonist.
[10d] The method according to any one of [10] to [10c], wherein the TNFR2 agonist is an anti-TNFR2 agonist antibody.
[10e] The method according to [10a], [10c] or [10d], wherein the TGF-βR agonist is TGF-β.
[10f] The method according to any one of [10] to [10e], wherein the culture in the step (1A) is further carried out in the presence of at least one agonist selected from the group consisting of a CD30 agonist, a CD3 agonist, and a CD28 agonist.
[10f1] The method according to any one of [10] to [10f], wherein in the culture of the step (1A), the culture is further performed in the presence of at least one agonist selected from the group consisting of a CD30 agonist, a CD3 agonist, and a CD28 agonist, and then in the absence of these agonists.
[10g] The method according to any one of [10] to [10f1], wherein the regulatory T cells are CD25 + /FOXP3 + cells.
[10h] The method according to [10g], wherein the CD25 + /FOXP3 + cells are CD4 + cells.
[10i] The method according to any one of [10] to [10f1], wherein the regulatory T cells are CD25 + /CD127 cells.
[10j] The method according to any one of [10] to [10i], wherein the regulatory T cells are Helios + cells.
[10k] (2) The method according to any one of [1] to [10j], comprising a step of introducing a foreign gene (particularly a gene for expressing a chimeric antigen receptor) into a cell population containing primary regulatory T cells or a cell population containing regulatory T cells obtained by the method according to any one of [1] to [10j].
[11] A cell population containing regulatory T cells obtained by the method according to any one of [1] to [10k].
[12] A pharmaceutical comprising a cell population containing regulatory T cells according to [11].
[13] The pharmaceutical agent according to [12], which is used for the prevention and/or treatment of abnormally enhanced immune response.
[14] A method for preventing and/or treating abnormally enhanced immune responses, comprising administering a cell population containing the regulatory T cells described in [11] to a subject in need thereof.
[15] A cell population comprising the regulatory T cells described in [11] for use in the prevention and/or treatment of abnormally enhanced immune responses.
[16] Use of a cell population containing regulatory T cells described in [11] in the manufacture of a pharmaceutical for preventing and/or treating abnormally enhanced immune responses.

 本発明によると、高い効率、かつ、その抑制機能を維持したまま、初代制御性T細胞を増殖させることが可能となる。 The present invention makes it possible to proliferate primary regulatory T cells with high efficiency while maintaining their suppressive function.

PBMCより初代培養Tregをフローサイトメータ―を用いて単離した結果を示す図である。ドットプロットの数値はTreg(CD4CD8αCD25CD127)の割合を示す。This shows the results of isolating primary cultured Tregs from PBMCs using a flow cytometer. The numbers in the dot plots indicate the percentage of Tregs (CD4 + CD8α CD25 + CD127 ). 初代培養TregをIL-2、IL-3、IL―4、IL-33、TGF-β1及び抗TNFR2抗体を含む培地で拡大培養をした際の経時的な細胞数の変化の結果を示すグラフである。Optimized:IL-2、IL-3、IL-4、IL-33、TGF-β1及び抗TNFR2抗体を含む培地、Control:IL-3、IL―4、IL-33、TGF-β1及び抗TNFR2抗体を含まない培地1 is a graph showing the change in cell number over time when primary cultured Tregs were expanded in a medium containing IL-2, IL-3, IL-4, IL-33, TGF-β1, and anti-TNFR2 antibody. Optimized: Medium containing IL-2, IL-3, IL-4, IL-33, TGF-β1, and anti-TNFR2 antibody. Control: Medium not containing IL-3, IL-4, IL-33, TGF-β1, or anti-TNFR2 antibody. IL-2、IL-3、IL-4、IL-33、TGF-β1及び抗TNFR2抗体を含む培地で拡大培養をしたTregに発現するタンパク質の発現量を検討した結果を示す図である。図における全てのドットプロットはCD3CD4CD8集団を示す。また、グラフの数値は各細胞集団におけるTreg(CD3CD4CD8CD25FOXP3)の割合を示す。This figure shows the results of examining the expression levels of proteins expressed in Tregs expanded in a medium containing IL-2, IL-3, IL-4, IL-33, TGF-β1, and an anti-TNFR2 antibody. All dot plots in the figure represent the CD3 + CD4 + CD8 population. The values on the graph indicate the proportion of Tregs (CD3 + CD4 + CD8 CD25 + FOXP3 + ) in each cell population. IL-2、IL-3、IL-4、IL-33、TGF-β1及び抗TNFR2抗体を含む培地で拡大培養をした細胞集団を用いて、ヒトPBMC由来の他家T細胞と共培養後のフローサイトメトリーによる分析結果を示す図(左)及びターゲット細胞の細胞分裂における抑制率(%)を示すグラフ(右)である。E:TはTregとヒトT細胞の細胞数の比を示す。The left figure shows the results of flow cytometry analysis of a cell population expanded in a medium containing IL-2, IL-3, IL-4, IL-33, TGF-β1, and an anti-TNFR2 antibody after co-culture with allogeneic T cells derived from human PBMCs, and the right graph shows the inhibition rate (%) of target cell division. E:T indicates the ratio of Treg to human T cell numbers. 初代培養TregをIL-2のみを含む培地、或いはIL-2、IL-3、IL-4、IL-33、TGF-β1及び抗TNFR2抗体を含む培地からIL-3、IL-4、IL-33及びTGF-β1のいずれか1つを抜いた培地で拡大培養をした際の経時的な細胞数の変化の結果を示すグラフである。FIG. 10 is a graph showing the change in cell number over time when primary cultured Tregs were expanded in a medium containing only IL-2, or in a medium containing IL-2, IL-3, IL-4, IL-33, TGF-β1, and an anti-TNFR2 antibody from which any one of IL-3, IL-4, IL-33, and TGF-β1 had been removed. IL-2、IL-3、IL-4、IL-33、TGF-β1及び抗TNFR2抗体を含む培地からIL-3、IL-4、IL-33及びTGF-β1のいずれか1つを抜いた培地で拡大培養をしたTregに発現するタンパク質の発現量を検討した結果を示す図である。図における全てのドットプロットはCD3CD4CD8集団を示す。また、グラフの数値は各細胞集団におけるTreg(CD3CD4CD8CD25FOXP3)の割合を示す。This figure shows the results of examining the expression levels of proteins expressed in Tregs expanded in a medium containing IL-2, IL-3, IL-4, IL-33, TGF-β1, and an anti-TNFR2 antibody, but omitting any one of IL-3, IL-4, IL-33, and TGF-β1. All dot plots in the figure represent the CD3 + CD4 + CD8 population. The values on the graph indicate the proportion of Tregs (CD3 + CD4 + CD8 CD25 + FOXP3 + ) in each cell population. IL-2、IL-3、IL-4、IL-33、TGF-β1及び抗TNFR2抗体を含む培地からIL-3、IL-4、IL-33及びTGF-β1のいずれか1つを抜いた培地で拡大培養をした細胞集団を用いて、ヒトPBMC由来の他家T細胞と共培養後のターゲット細胞の細胞分裂における抑制率(%)を示すグラフである。E:TはTregとヒトT細胞の細胞数の比を示す。1 is a graph showing the rate (%) of inhibition of target cell division after co-culture with human PBMC-derived allogeneic T cells using a cell population expanded in a medium containing IL-2, IL-3, IL-4, IL-33, TGF-β1, and an anti-TNFR2 antibody but from which any one of IL-3, IL-4, IL-33, and TGF-β1 has been removed. E:T indicates the ratio of the number of Tregs to the number of human T cells. 初代培養TregをIL-2、IL-3、IL-4、IL-33、TGF-β1及び抗TNFR2抗体を含む培地、或いは、上記培地組成からIL-3、TGF-β1、及び抗TNFR2抗体を抜いた培地で拡大培養をした際の経時的な細胞数の変化の結果を示すグラフである。FIG. 1 is a graph showing the change in cell number over time when primary cultured Tregs were expanded in a medium containing IL-2, IL-3, IL-4, IL-33, TGF-β1, and an anti-TNFR2 antibody, or in a medium containing the above medium composition minus IL-3, TGF-β1, and anti-TNFR2 antibody.

 以下、本発明の実施の形態について詳細に説明する。 The following describes in detail the embodiments of the present invention.

 「~を含む(comprise(s)又はcomprising)」とは、その語句に続く要素の包含を示すがこれに限定されないことを意味する。したがって、その語句に続く要素の包含は示唆するが、他の任意の要素の除外は示唆しない。「~からなる(consist(s) of又はconsisting of)」とは、その語句に続くあらゆる要素を包含し、かつ、これに限定されることを意味する。したがって、「~からなる」という語句は、列挙された要素が要求されるか又は必須であり、他の要素は実質的に存在しないことを示す。「~から本質的になる(consist(s) essentially of又はconsisting essentially of)」とは、その語句に続く任意の要素を包含し、かつ、その要素について本開示で特定された活性又は作用に影響しない他の要素に限定されることを意味する。したがって、「~から本質的になる」という語句は、列挙された要素が要求されるか又は必須であるが、他の要素は任意選択であり、それらが列挙された要素の活性又は作用に影響を及ぼすかどうかに応じて、存在させる場合もあり、存在させない場合もあることを示す。 "Comprise(s)" or "comprising" means the inclusion of, but is not limited to, the elements that follow the phrase. Thus, it implies the inclusion of the elements that follow the phrase, but does not imply the exclusion of any other elements. "Consist(s) of" means inclusive of and limited to all elements that follow the phrase. Thus, the phrase "consisting of" indicates that the listed elements are required or essential, and that other elements are substantially absent. "Consist(s) essentially of" means inclusive of any elements that follow the phrase, and is limited to other elements that do not affect the activity or function of the element identified in this disclosure. Thus, the phrase "consisting essentially of" indicates that the listed elements are required or essential, but that other elements are optional and may or may not be present depending on whether they affect the activity or function of the listed elements.

 本明細書において、「培養」とは、細胞をインビトロ環境において維持し、又は/及び増殖させることを指す。「培養する」とは、組織外又は体外で、例えば、細胞培養ディッシュ又はフラスコ中で細胞を持続させ、又は/及び増殖させることを意味する。 As used herein, "culture" refers to maintaining and/or growing cells in an in vitro environment. "Culturing" refers to maintaining and/or growing cells outside a tissue or outside the body, for example, in a cell culture dish or flask.

 本明細書において、「物質の存在下で細胞集団を培養する」とは、例えば、物質を含む培地で細胞集団を培養することを指す。そのような培養としては、例えば、当該物質のみ、又は該物質と他の分化誘導因子などとが共存した培地での培養を挙げることができる。当該物質を培地に添加する場合には、培地に直接添加するか、もしくは適切な溶媒にて当該物質を用時溶解した後、培地中に添加してもよい。また、当該物質を培養の際の基板及び担体表面上に固定化して培養することもできる。 As used herein, "culturing a cell population in the presence of a substance" refers to culturing a cell population in a medium containing the substance. Examples of such culturing include culturing in a medium containing the substance alone, or in which the substance coexists with other differentiation-inducing factors. When adding the substance to the medium, it can be added directly to the medium, or the substance can be dissolved in an appropriate solvent just before use and then added to the medium. The substance can also be immobilized on the surface of a substrate or carrier during culturing and then cultured.

 本明細書において、「陽性(positive(+))」とは、タンパク質又は遺伝子が当該分野で公知の手法による検出可能量で発現していることを意味する。また、細胞内に発現し、細胞表面には現れないタンパク質(例えば転写因子又はそのサブユニットなど)の場合は、当該タンパク質とともにレポータータンパク質を発現させ、当該レポータータンパク質を検出することによって対象とするタンパク質を検出できる。遺伝子の検出は、例えば、RT-PCR、マイクロアレイ、バイオチップ及びRNAseq等の核酸増幅方法及び/又は核酸検出方法を利用して行うことができる。 As used herein, "positive (+)" means that the protein or gene is expressed in a detectable amount using techniques known in the art. In addition, in the case of proteins that are expressed intracellularly and not on the cell surface (e.g., transcription factors or their subunits), the target protein can be detected by expressing a reporter protein along with the protein and detecting the reporter protein. Gene detection can be performed using nucleic acid amplification and/or nucleic acid detection methods, such as RT-PCR, microarrays, biochips, and RNAseq.

 本明細書において、「陰性(negative(-))」とは、タンパク質又は遺伝子の発現量が、上記のような公知手法の全てあるいはいずれかによる検出下限値未満であること、あるいはその発現の程度が低発現性であることを意味する。タンパク質又は遺伝子の発現の検出下限値は、各手法により異なりえる。タンパク質又は遺伝子の発現の程度(低発現性であるか、高発現性であるか)は、同一条件で測定した対照細胞の結果との比較により、判断することができる。例えば、ある細胞の集団におけるCD25の発現の程度は、フローサイトメトリーを用い、その細胞集団におけるCD25発現量を、PBMC由来Tconv(対照:CD25低発現性であることが知られている。)におけるCD25発現量と比較し、対照と同等の発現が見られる場合は低発現性であると判断し、対照細胞より発現が高い場合は高発現性であると判断することができる。 As used herein, "negative (-)" means that the expression level of a protein or gene is below the lower limit of detection by any or all of the above-mentioned known techniques, or that the level of expression is low. The lower limit of detection for protein or gene expression may vary depending on the technique. The level of protein or gene expression (whether low or high) can be determined by comparing it with the results of control cells measured under the same conditions. For example, the level of CD25 expression in a certain cell population can be determined using flow cytometry, by comparing the CD25 expression level in that cell population with the CD25 expression level in PBMC-derived Tconv (control: known to be a low CD25 expressor). If expression equivalent to that of the control is observed, it is determined to be low expression, and if expression is higher than that of the control cells, it is determined to be high expression.

 本明細書において、「マーカー」とは、所定の細胞型において細胞表面、細胞質内、核内等に特異的に発現されるタンパク質又はその遺伝子を意味する。マーカーは、好ましくは「細胞表面マーカー」である。「細胞表面マーカー」とは、蛍光物質により標識(染色)可能であり、細胞表面マーカーを発現している細胞の検出、濃縮、単離等を容易に行える、細胞表面に発現するタンパク質を指す。当該細胞表面マーカーとは、所定の細胞型において特異的に発現する(陽性マーカー)又は発現しない(陰性マーカー)遺伝子、具体的にはゲノム中の当該遺伝子の転写によるmRNAとして、又はそのmRNAの翻訳によるタンパク質として、生成する(陽性マーカー)又は生成しない(陰性マーカー)物質を指す。 As used herein, the term "marker" refers to a protein or its gene that is specifically expressed on the cell surface, in the cytoplasm, in the nucleus, etc., of a specific cell type. The marker is preferably a "cell surface marker." A "cell surface marker" refers to a protein expressed on the cell surface that can be labeled (stained) with a fluorescent substance, making it easy to detect, concentrate, isolate, etc., cells expressing the cell surface marker. The cell surface marker refers to a gene that is specifically expressed (positive marker) or not expressed (negative marker) in a specific cell type; specifically, a substance that is produced (positive marker) or not produced (negative marker) as mRNA resulting from transcription of the gene in the genome, or as a protein resulting from translation of that mRNA.

 このような細胞表面マーカーの検出は、当該細胞表面マーカーに特異的な抗体を用いた免疫学的アッセイ、例えば、ELISA、免疫染色、フローサイトメトリーを利用して行うことができる。 Such cell surface markers can be detected using immunological assays using antibodies specific to the cell surface markers, such as ELISA, immunostaining, and flow cytometry.

 本明細書において、「発現(expression)」とは、細胞内のプロモーターにより駆動される特定のヌクレオチド配列の転写及び/又は翻訳として定義される。 As used herein, "expression" is defined as the transcription and/or translation of a specific nucleotide sequence driven by a promoter within a cell.

 本明細書において、「制御性T細胞」とは、T細胞受容体を介する刺激を受けたときに、エフェクターT細胞の活性化を阻害し得る能力を有し、免疫応答の抑制(免疫寛容)を司っているT細胞を意味する。制御性T細胞は、通常CD25/FOXP3細胞又はCD25/CD127細胞であり、その中に、CD4又はCD8の細胞が存在する。本発明において、制御性T細胞は、CD4の細胞(すなわち、CD4/CD25/FOXP3又はCD4/CD25/CD127)であってもよいし、CD8の細胞(すなわち、CD8/CD25/FOXP3又はCD8/CD25/CD127)であってもよく、CD4の細胞がより好ましい。また、CD4/CD25制御性T細胞のマスターレギュレーターとして、転写因子FOXP3が知られている。その他、制御性T細胞は、制御性T細胞の抑制機能を示す指標として知られている、Helios、CTLA4(Cytotoxic T Lymphocyte (associated) Antigen 4)(CD152)、CD39及びCD73が陽性であり得る。Heliosは、Ikaros転写因子ファミリーのメンバーであり、FOXP3プロモーターに結合し、FOXP3の発現を増加させることが示されている。CTLA4は免疫チェックポイント・タンパク質であり、T細胞の表面に発現したCTLA4が抗原提示細胞の表面でCD80又はCD86に結合すると、T細胞の活性化が抑制される。また、CD39及びCD73はそれぞれATPとAMPを加水分解して最終産物としてアデノシンを産生し、アデノシンがT細胞に作用するとT細胞の活性化が抑制されるというメカニズムが知られている。なお、本明細書中、「CD25/FOXP3」などの表記において用いられる「/」とは「及び」を意味する。 As used herein, the term "regulatory T cells" refers to T cells that, upon stimulation via a T cell receptor, have the ability to inhibit the activation of effector T cells and are responsible for suppressing immune responses (immune tolerance). Regulatory T cells are usually CD25 + /FOXP3 + cells or CD25 + /CD127 - cells, among which CD4 + or CD8 + cells are present. In the present invention, regulatory T cells may be CD4 + cells (i.e., CD4 + /CD25 + /FOXP3 + or CD4 + /CD25 + /CD127 - ) or CD8 + cells (i.e., CD8 + /CD25 + /FOXP3 + or CD8 + /CD25 + /CD127 - ), with CD4 + cells being more preferred. Furthermore, the transcription factor FOXP3 is known as a master regulator of CD4 + /CD25 + regulatory T cells. Regulatory T cells may also be positive for Helios, CTLA4 (Cytotoxic T Lymphocyte (associated) Antigen 4) (CD152), CD39, and CD73, which are known to be indicators of the suppressive function of regulatory T cells. Helios is a member of the Ikaros transcription factor family and has been shown to bind to the FOXP3 promoter and increase FOXP3 expression. CTLA4 is an immune checkpoint protein, and when CTLA4 expressed on the surface of T cells binds to CD80 or CD86 on the surface of antigen-presenting cells, T cell activation is suppressed. It is also known that CD39 and CD73 hydrolyze ATP and AMP, respectively, to produce adenosine as the final product, and that the activation of T cells is suppressed when adenosine acts on T cells. In this specification, the "/" used in expressions such as "CD25 + /FOXP3 + " means "and."

 本明細書において、「細胞集団(population)」とは、同じ種類又は異なる種類の2以上の細胞を意味する。「細胞集団(population)」は、同じ種類又は異なる種類の細胞の一塊(mass)をも意味する。 As used herein, "cell population" refers to two or more cells of the same or different types. "Cell population" also refers to a mass of cells of the same or different types.

 本明細書において、「制御性T細胞が増殖した」、「制御性T細胞の増殖」とは、細胞集団における制御性T細胞の数(絶対数)が、培養前、又は本発明を実施せずに得られた細胞のような対照と比較して増加していることを指し、例えば、培養前又は対照と比較して少なくとも1.1倍、1.2倍、1.5倍、2倍、3倍、4倍、5倍、6倍、7倍、8倍、9倍、10倍、15倍、20倍、30倍、40倍、50倍、100倍、300倍、500倍、1000倍、2000倍、3000倍、4000倍、5000倍、6000倍、7000倍、8000倍、9000倍、10000倍、15000倍、20000倍、50000倍、100000倍増加させることを意味する。本発明の特定の実施形態では、本発明により製造される細胞集団における制御性T細胞の数(絶対数)は、培養前又は対照の細胞の数と比較して、少なくとも1.1倍、1.2倍、1.5倍、2倍、3倍、4倍、5倍、6倍、7倍、8倍、9倍、10倍、15倍、20倍、30倍、40倍、50倍、100倍、300倍、500倍、1000倍、2000倍、3000倍、4000倍、5000倍、6000倍、7000倍、8000倍、9000倍、10000倍、15000倍、20000倍、50000倍、100000倍増加するよう、製造される。 As used herein, "regulatory T cells expanded" and "expansion of regulatory T cells" refer to an increase in the number (absolute number) of regulatory T cells in a cell population compared to before culture or a control such as cells obtained without carrying out the present invention, and mean, for example, an increase of at least 1.1-fold, 1.2-fold, 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 300-fold, 500-fold, 1000-fold, 2000-fold, 3000-fold, 4000-fold, 5000-fold, 6000-fold, 7000-fold, 8000-fold, 9000-fold, 10000-fold, 15000-fold, 20000-fold, 50000-fold, or 100000-fold compared to before culture or the control. In certain embodiments of the present invention, the number (absolute number) of regulatory T cells in a cell population produced by the present invention is increased by at least 1.1-fold, 1.2-fold, 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 300-fold, 500-fold, 1000-fold, 2000-fold, 3000-fold, 4000-fold, 5000-fold, 6000-fold, 7000-fold, 8000-fold, 9000-fold, 10000-fold, 15000-fold, 20000-fold, 50000-fold, or 100000-fold compared to the number of cells before culture or in a control.

 本発明で用いる各種の細胞は、治療への適用の観点からは、GMP(Good Manufacturing Practice)規格の細胞であることが好ましい。 From the perspective of therapeutic application, it is preferable that the various cells used in the present invention are GMP (Good Manufacturing Practice) standard cells.

 「核酸」とは、ヌクレオチド及び該ヌクレオチドと同等の機能を有する分子が重合した分子であればいかなるものでもよく、例えば、リボヌクレオチドの重合体であるRNA、デオキシリボヌクレオチドの重合体であるDNA、リボヌクレオチド及びデオキシリボヌクレオチドが混合した重合体、及びヌクレオチド類似体を含むヌクレオチド重合体を挙げることができ、さらに、核酸誘導体を含むヌクレオチド重合体であってもよい。核酸は、一本鎖核酸又は二本鎖核酸であってもよい。二本鎖核酸には、一方の鎖に対し、他方の鎖がストリンジェントな条件でハイブリダイズする二本鎖核酸も含まれる。 "Nucleic acid" refers to any molecule formed by polymerizing nucleotides and molecules with equivalent functions to the nucleotides. Examples include RNA, which is a polymer of ribonucleotides; DNA, which is a polymer of deoxyribonucleotides; mixed polymers of ribonucleotides and deoxyribonucleotides; and nucleotide polymers containing nucleotide analogs. Nucleic acids may also be nucleotide polymers containing nucleic acid derivatives. Nucleic acids may be single-stranded or double-stranded. Double-stranded nucleic acids also include double-stranded nucleic acids in which one strand hybridizes to the other strand under stringent conditions.

 ヌクレオチド類似体としては、RNA又はDNAと比較して、ヌクレアーゼ耐性の向上又は、安定化させるため、相補鎖核酸とのアフィニティーを上げるため、あるいは細胞透過性を上げるため、あるいは可視化させるために、リボヌクレオチド、デオキシリボヌクレオチド、RNA又はDNAに修飾を施した分子であればいかなる分子でもよい。ヌクレオチド類似体としては、天然に存在する分子でも非天然の分子でもよく、例えば、糖部修飾ヌクレオチド類似体(例:2’-O-メチルリボースで置換されたヌクレオチド類似体、2’-O-プロピルリボースで置換されたヌクレオチド類似体、2’-メトキシエトキシリボースで置換されたヌクレオチド類似体、2’-O-メトキシエチルリボースで置換されたヌクレオチド類似体、2’-O-[2-(グアニジウム)エチル]リボースで置換されたヌクレオチド類似体、2’-フルオロリボースで置換されたヌクレオチド類似体、架橋構造型人工核酸(BNA:Bridged Nucleic Acid)、ロックド人工核酸(LNA:Locked Nucleic Acid)、エチレン架橋構造型人工核酸(ENA:Ethylene bridged nucleic acid)、ペプチド核酸(PNA)、オキシペプチド核酸(OPNA)、ペプチドリボ核酸(PRNA))、リン酸ジエステル結合修飾ヌクレオチド類似体(例:ホスフォロチオエート結合に置換されたヌクレオチド類似体、N3’-P5’ホスフォアミデート結合に置換されたヌクレオチド類似体)等が挙げられる。 Nucleotide analogs may be any molecule in which ribonucleotides, deoxyribonucleotides, RNA, or DNA have been modified to improve nuclease resistance or stabilization compared to RNA or DNA, increase affinity with complementary nucleic acids, increase cell permeability, or enable visualization. Nucleotide analogs may be naturally occurring or unnatural molecules, such as sugar-modified nucleotide analogs (e.g., nucleotide analogs substituted with 2'-O-methylribose, nucleotide analogs substituted with 2'-O-propylribose, nucleotide analogs substituted with 2'-methoxyethoxyribose, nucleotide analogs substituted with 2'-O-methoxyethylribose, nucleotide analogs substituted with 2'-O-[2-(guanidinium)ethyl]ribose, nucleotide analogs substituted with 2'-fluororibose, and bridged artificial nucleotides. Examples include nucleic acids (BNA: Bridged Nucleic Acid), locked artificial nucleic acids (LNA: Locked Nucleic Acid), ethylene-bridged artificial nucleic acids (ENA: Ethylene-bridged nucleic acid), peptide nucleic acids (PNA), oxypeptide nucleic acids (OPNA), peptide ribonucleic acids (PRNA), nucleotide analogs modified with phosphodiester bonds (e.g., nucleotide analogs substituted with phosphorothioate bonds, nucleotide analogs substituted with N3'-P5' phosphoamidate bonds), etc.

 核酸誘導体としては、核酸に比べ、ヌクレアーゼ耐性を向上させるため、安定化させるため、相補鎖核酸とのアフィニティーを上げるため、細胞透過性を上げるため、あるいは可視化させるために、該核酸に別の化学物質を付加した分子であればいかなる分子でもよく、その具体例としては、5’-ポリアミン付加誘導体、コレステロール付加誘導体、ステロイド付加誘導体、胆汁酸付加誘導体、ビタミン付加誘導体、Cy5付加誘導体、Cy3付加誘導体、6-FAM付加誘導体、ビオチン付加誘導体等を挙げることができる。 A nucleic acid derivative can be any molecule in which another chemical substance has been added to the nucleic acid in order to improve nuclease resistance, stabilization, affinity with complementary nucleic acid strands, cell permeability, or visualization, compared to nucleic acids. Specific examples include 5'-polyamine-added derivatives, cholesterol-added derivatives, steroid-added derivatives, bile acid-added derivatives, vitamin-added derivatives, Cy5-added derivatives, Cy3-added derivatives, 6-FAM-added derivatives, and biotin-added derivatives.

 本発明の制御性T細胞が増殖した細胞集団の製造方法(本明細書において、単に「本発明の製造方法」と称することがある)は、以下の工程を含むことを特徴とする。
(1)インターロイキン(IL)-2、IL-4、IL-33及びTNFR2アゴニストの存在下で初代制御性T細胞を含む細胞集団を培養する工程。
The method of the present invention for producing a cell population in which regulatory T cells have expanded (sometimes referred to herein simply as the "production method of the present invention") is characterized by comprising the following steps:
(1) Culturing a cell population containing primary regulatory T cells in the presence of interleukin (IL)-2, IL-4, IL-33, and a TNFR2 agonist.

 工程(1)は、IL-2、IL-4、IL-33及びTNFR2アゴニストに加えて、TGF-βRアゴニスト及び/又はIL-3の存在下で行うこともできる。すなわち、工程(1)は、IL-2、IL-3、IL-4、IL-33及びTNFR2アゴニストの存在下、IL-2、IL-4、IL-33、TGF-βRアゴニスト及びTNFR2アゴニストの存在下、又はIL-2、IL-3、IL-4、IL-33、TGF-βRアゴニスト及びTNFR2アゴニストの存在下などで行うこともできる。なお、工程(1)は、制御性T細胞の増殖の観点から、mTOR阻害剤(例えば、ラパマイシン)の非存在下で行うことが好ましい。 Step (1) can also be carried out in the presence of a TGF-βR agonist and/or IL-3 in addition to IL-2, IL-4, IL-33, and a TNFR2 agonist. That is, step (1) can be carried out in the presence of IL-2, IL-3, IL-4, IL-33, and a TNFR2 agonist; in the presence of IL-2, IL-4, IL-33, a TGF-βR agonist, and a TNFR2 agonist; or in the presence of IL-2, IL-3, IL-4, IL-33, a TGF-βR agonist, and a TNFR2 agonist. From the perspective of regulatory T cell proliferation, step (1) is preferably carried out in the absence of an mTOR inhibitor (e.g., rapamycin).

 IL(インターロイキン)-4、IL(インターロイキン)-3、IL(インターロイキン)-2、及びIL(インターロイキン)-33は、哺乳類由来が好ましく、特にヒト由来が好ましい。また、IL-2、IL-3、IL-4、及びIL-33は、哺乳動物(特にヒト)から単離及び精製された天然のものであってもよく、又は遺伝子工学的手法により人工的に製造されたもの(アミノ酸の置換、欠失、挿入及び/又は付加などが施されていてもよい)であってもよい。 IL-4, IL-3, IL-2, and IL-33 are preferably derived from mammals, and particularly preferably from humans. IL-2, IL-3, IL-4, and IL-33 may be natural products isolated and purified from mammals (particularly humans), or may be artificially produced using genetic engineering techniques (which may include amino acid substitutions, deletions, insertions, and/or additions).

 TGF-βR(Transforming Growth Factor-β Receptor)アゴニストとは、TGF-βRに結合して、TGF-βRのシグナル伝達を活性化することができる物質と定義される。TGF-βRアゴニストとしては、TGF-βスーパーファミリーに属する因子が挙げられ、TGF-βスーパーファミリーにはTGF-βファミリー、アクチビンファミリー、及びBMPファミリー(bone morphogenetic protein)が存在する。そのようなTGF-βスーパーファミリーに属する因子としては、例えば、TGF-β(TGF-β1、TGF-β2、TGF-β3)、アクチビンA、アクチビンB、GDF-8、GDF-11などが挙げられる。TGF-βRアゴニストとしては、好ましくはTGF-β、より好ましくはTGF-β1である。TGF-βRアゴニストは、1種類単独で、又は2種類以上を組み合わせて使用することができる。 A TGF-βR (Transforming Growth Factor-β Receptor) agonist is defined as a substance that can bind to TGF-βR and activate TGF-βR signal transduction. Examples of TGF-βR agonists include factors belonging to the TGF-β superfamily, which includes the TGF-β family, the activin family, and the BMP (bone morphogenetic protein) family. Factors belonging to the TGF-β superfamily include, for example, TGF-β (TGF-β1, TGF-β2, TGF-β3), activin A, activin B, GDF-8, and GDF-11. A preferred TGF-βR agonist is TGF-β, and more preferably TGF-β1. TGF-βR agonists can be used singly or in combination of two or more.

 TNFR2(Tumor necrosis Factor Receptor-2)アゴニストとは、TNFR2に結合して、TNFR2のシグナル伝達を活性化することができる物質と定義され、例えば、抗体(例えば、抗TNFR2抗体)、ペプチド、低分子化合物、タンパク質などである。抗TNFR2アゴニスト抗体としては、例えば、クローンMR2-1(Hycult Biotech社)、クローンMAB2261(R&D Systems社)などのTNFR2に結合するモノクローナル抗体などが挙げられる。TNFR2アゴニストはまた、アゴニストとしてTNFR2にのみ結合するTNF-αムテインであってもよい。TNFR2アゴニストは、1種類単独で、又は2種類以上を組み合わせて使用することができる。 TNFR2 (Tumor Necrosis Factor Receptor-2) agonists are defined as substances that can bind to TNFR2 and activate TNFR2 signaling, and include, for example, antibodies (e.g., anti-TNFR2 antibodies), peptides, small molecules, and proteins. Examples of anti-TNFR2 agonist antibodies include monoclonal antibodies that bind to TNFR2, such as clone MR2-1 (Hycult Biotech) and clone MAB2261 (R&D Systems). TNFR2 agonists may also be TNF-α muteins that bind only to TNFR2 as agonists. TNFR2 agonists can be used singly or in combination of two or more.

 TNFR2アゴニストとしては、好ましくは抗TNFR2アゴニスト抗体である。当該抗体としては、その機能性断片であってもよく、機能性断片としては、例えば、Fd、Fv、Fab、F(ab’)、F(ab)、F(ab’)、単鎖Fv(scFv)、ダイアボディ、トリアボディ(triabody)、テトラボディ(tetrabody)及びミニボディを挙げることができる。抗体としては、マウス、ラット、ウシ、ウサギ、ヤギ、ヒツジ、モルモット等の動物由来のものが挙げられる。抗体のアイソタイプは特に制限されず、アイソタイプとしては、例えば、IgG(IgG1、IgG2、IgG3、IgG4)、IgA、IgD、IgE及びIgMが挙げられる。また、抗体は、モノクローナル抗体及びポリクローナル抗体のいずれであってもよく、好ましくはモノクローナル抗体であり、また、抗体は、ヒト化抗体、キメラ抗体、多重特異性抗体(例えば、二重特異性抗体)などであってもよい。抗体は、公知の方法により製造することが可能であり、例えば、抗体をコードする核酸を含有する発現ベクターを構築し、当該核酸を導入した形質転換体を培養すること、抗体を産生するハイブリドーマを培養することなどにより生産することができる。 The TNFR2 agonist is preferably an anti-TNFR2 agonist antibody. The antibody may be a functional fragment thereof, such as Fd, Fv, Fab, F(ab'), F(ab) 2 , F(ab') 2 , single-chain Fv (scFv), diabody, triabody, tetrabody, and minibody. Antibodies include those derived from animals such as mouse, rat, cow, rabbit, goat, sheep, and guinea pig. The antibody isotype is not particularly limited, and examples of isotypes include IgG (IgG1, IgG2, IgG3, IgG4), IgA, IgD, IgE, and IgM. Furthermore, the antibody may be either a monoclonal antibody or a polyclonal antibody, preferably a monoclonal antibody, and may also be a humanized antibody, a chimeric antibody, a multispecific antibody (e.g., a bispecific antibody), etc. Antibodies can be produced by known methods, for example, by constructing an expression vector containing a nucleic acid encoding the antibody, culturing a transformant into which the nucleic acid has been introduced, or culturing a hybridoma that produces the antibody.

 また、IL-2、IL-3、IL-4、IL-33、TGF-βRアゴニスト及びTNFR2アゴニストは、フリーの状態又は塩の状態で使用することができる。塩としては、例えば、ナトリウム塩、マグネシウム塩、カリウム塩、カルシウム塩、アルミニウム塩等の無機塩基との塩;メチルアミン塩、エチルアミン塩、エタノールアミン塩等の有機塩基との塩;リジン、オルニチン、アルギニン等の塩基性アミノ酸との塩及びアンモニウム塩が挙げられる。当該塩は、酸付加塩であってもよく、かかる塩としては、具体的には、塩酸、臭化水素酸、ヨウ化水素酸、硫酸、硝酸、リン酸等の鉱酸;ギ酸、酢酸、プロピオン酸、シュウ酸、マロン酸、リンゴ酸、酒石酸、フマル酸、コハク酸、乳酸、マレイン酸、クエン酸、メタンスルホン酸、エタンスルホン酸等の有機酸;アスパラギン酸、グルタミン酸等の酸性アミノ酸との酸付加塩が挙げられる。IL-2、IL-3、IL-4、IL-33、TGF-βRアゴニスト及びTNFR2アゴニストには、水和物、溶媒和物、結晶多形なども含まれる。 IL-2, IL-3, IL-4, IL-33, TGF-βR agonists, and TNFR2 agonists can be used in their free state or in the form of a salt. Examples of salts include salts with inorganic bases such as sodium salt, magnesium salt, potassium salt, calcium salt, and aluminum salt; salts with organic bases such as methylamine salt, ethylamine salt, and ethanolamine salt; salts with basic amino acids such as lysine, ornithine, and arginine; and ammonium salts. The salts may also be acid addition salts, and specific examples of such salts include acid addition salts with mineral acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, nitric acid, and phosphoric acid; organic acids such as formic acid, acetic acid, propionic acid, oxalic acid, malonic acid, malic acid, tartaric acid, fumaric acid, succinic acid, lactic acid, maleic acid, citric acid, methanesulfonic acid, and ethanesulfonic acid; and acidic amino acids such as aspartic acid and glutamic acid. IL-2, IL-3, IL-4, IL-33, TGF-βR agonists, and TNFR2 agonists also include hydrates, solvates, and crystalline polymorphs.

 培地中におけるIL-2の濃度は、特に限定されず、例えば、0.1~1000ng/mL、好ましくは10~500ng/mL、より好ましくは50~200ng/mLである。 The concentration of IL-2 in the culture medium is not particularly limited, and is, for example, 0.1 to 1000 ng/mL, preferably 10 to 500 ng/mL, and more preferably 50 to 200 ng/mL.

 培地中におけるIL-4の濃度は、特に限定されず、例えば、0.01~100ng/mL、好ましくは10~100ng/mLである。 The concentration of IL-4 in the culture medium is not particularly limited, and is, for example, 0.01 to 100 ng/mL, preferably 10 to 100 ng/mL.

 培地中におけるIL-33の濃度は、特に限定されず、例えば、0.1~100ng/mL、好ましくは10~100ng/mLである。 The concentration of IL-33 in the culture medium is not particularly limited, and is, for example, 0.1 to 100 ng/mL, preferably 10 to 100 ng/mL.

 培地中におけるTNFR2アゴニストの濃度は、特に限定されず、用いるTNFR2アゴニストの種類などによって適宜調整される。TNFR2アゴニストの濃度は、例えば、0.0001~100μg/mL、好ましくは0.01~10μg/mLである。 The concentration of the TNFR2 agonist in the medium is not particularly limited and can be adjusted appropriately depending on the type of TNFR2 agonist used. The concentration of the TNFR2 agonist is, for example, 0.0001 to 100 μg/mL, preferably 0.01 to 10 μg/mL.

 培地中におけるTGF-βRアゴニストの濃度は、特に限定されず、用いるTGF-βRアゴニストの種類などによって適宜調整される。TGF-βRアゴニストの濃度は、例えば、0.1~100ng/mL、好ましくは1~50ng/mLである。 The concentration of the TGF-βR agonist in the culture medium is not particularly limited and can be adjusted appropriately depending on the type of TGF-βR agonist used. The concentration of the TGF-βR agonist is, for example, 0.1 to 100 ng/mL, preferably 1 to 50 ng/mL.

 培地中におけるIL-3の濃度は、特に限定されず、例えば、0.01~100ng/mL、好ましくは10~100ng/mLである。 The concentration of IL-3 in the culture medium is not particularly limited, and is, for example, 0.01 to 100 ng/mL, preferably 10 to 100 ng/mL.

 工程(1)の培養で使用される培地は、動物細胞の培養に用いられる培地を基礎培地として、前述するIL-2、IL-4、IL-33、TNFR2アゴニスト(及びIL-3、TGF-βRアゴニスト等)を含むものである。基礎培地としては、動物細胞の培養に使用できるものであれば特に限定されず、例えば、AIM V、X-VIVO-15、NeuroBasal、EGM2、TeSR、BME、BGJb、CMRL 1066、グラスゴーMEM、改良MEM亜鉛オプション、IMDM、199培地、イーグルMEM、αMEM、DMEM、Ham、RPMI-1640、フィッシャー培地などが挙げられる。これらの培地は、いずれか1種を単独で用いてもよいし、2種以上を混合して用いてもよい。 The medium used in the culture of step (1) is a basal medium that is used for culturing animal cells and contains the aforementioned IL-2, IL-4, IL-33, and TNFR2 agonist (as well as IL-3, TGF-βR agonist, etc.). The basal medium is not particularly limited as long as it can be used for culturing animal cells, and examples include AIM V, X-VIVO-15, NeuroBasal, EGM2, TeSR, BME, BGJb, CMRL 1066, Glasgow MEM, Improved MEM Zinc Option, IMDM, 199 Medium, Eagle's MEM, αMEM, DMEM, Ham, RPMI-1640, and Fischer's Medium. Any one of these media may be used alone, or two or more may be used in combination.

 培地は、血清を含有していてもよいし、無血清でもよい。培地はまた、血清代替物(例えば、アルブミン、トランスフェリン、Knockout Serum Replacement(KSR)、脂肪酸、インスリン、コラーゲン前駆体、微量元素、2-メルカプトエタノール、3’-チオールグリセロール、ITS-サプリメント、B27(商標)サプリメント等)を含有していてもよい。血清代替物は、1種又は2種以上を使用することができる。 The medium may contain serum or may be serum-free. The medium may also contain serum substitutes (e.g., albumin, transferrin, Knockout Serum Replacement (KSR), fatty acids, insulin, collagen precursors, trace elements, 2-mercaptoethanol, 3'-thiolglycerol, ITS supplement, B27™ supplement, etc.). One or more types of serum substitutes may be used.

 さらに、培地には、脂質、アミノ酸(非必須アミノ酸等)、L-グルタミン、ビタミン、増殖因子、サイトカイン、抗CD3抗体、抗CD30抗体、抗CD28抗体、抗生物質、抗酸化剤、ピルビン酸、緩衝剤、無機塩類などの1つ以上の物質も含有することができる。培地としては、血清等の成分が明らかでないものを含まない既知組成(chemically-defined)の培地を使用することが、培地のロット間の差が低減され、品質の安定した細胞を調製できるため望ましい。 Furthermore, the medium may contain one or more substances such as lipids, amino acids (non-essential amino acids, etc.), L-glutamine, vitamins, growth factors, cytokines, anti-CD3 antibodies, anti-CD30 antibodies, anti-CD28 antibodies, antibiotics, antioxidants, pyruvic acid, buffers, and inorganic salts. It is preferable to use a chemically defined medium that does not contain unknown components such as serum, as this reduces differences between medium lots and allows for the preparation of cells of consistent quality.

 培地のpHは、通常7.0~7.8、好ましくは7.2~7.6である。培地は、使用前にはコンタミネーションを防止するため、濾過、紫外線照射、加熱滅菌、放射線照射等の方法により好ましくは滅菌される。 The pH of the medium is usually 7.0 to 7.8, preferably 7.2 to 7.6. Prior to use, the medium is preferably sterilized by filtration, ultraviolet irradiation, heat sterilization, radiation, or other methods to prevent contamination.

 培養は、フィーダー細胞の存在下又は非存在下で実施される。本発明の製造方法は、成分が明らかでないものが混入することがなく、均一な性質を持つ制御性T細胞を安定して製造させることができるため、フィーダー細胞の非存在下で実施されることが望ましい。 Culturing can be carried out in the presence or absence of feeder cells. The production method of the present invention is preferably carried out in the absence of feeder cells, as it allows for the stable production of regulatory T cells with uniform properties without contamination by unknown components.

 本発明の製造方法の培養条件は、特に制限されず、培養温度は、例えば、37~42℃程度、好ましくは37~39℃程度であり、CO濃度は、例えば2~10%、好ましくは2~5%であり、酸素濃度は、例えば1~20%、好ましくは5~20%である。 The culture conditions for the production method of the present invention are not particularly limited. The culture temperature is, for example, about 37 to 42°C, preferably about 37 to 39°C, the CO2 concentration is, for example, 2 to 10%, preferably 2 to 5%, and the oxygen concentration is, for example, 1 to 20%, preferably 5 to 20%.

 また、培養期間についても、特に制限されず、当業者であれば制御性T細胞の数などをモニターしながら適宜決定することが可能であり、例えば、7日間以上、好ましくは14日間以上、より好ましくは21日以上、更に好ましくは28日以上である。培養期間の上限は特に限定されず、例えば、42日間以下、好ましくは35日間以下、より好ましくは28日間以下である。本発明における培養では、所望の量の制御性T細胞を得るために必要な回数継代を行ってもよいし、培地の追加及び交換を行ってもよい。本発明における培養は、公知のCOインキュベーターを使用して行うことができる。培養容器は特に限定されるものではなく、プレート、ディッシュ、シャーレ、フラスコ、バッグ、ボトル、タンク(培養槽)、バイオリアクターなどの中から適宜選択することができる。培養容器としては抗CD3抗体を結合させた容器を用いることができる。 The culture period is also not particularly limited and can be appropriately determined by those skilled in the art while monitoring the number of regulatory T cells, etc., and is, for example, 7 days or more, preferably 14 days or more, more preferably 21 days or more, and even more preferably 28 days or more. The upper limit of the culture period is not particularly limited and, for example, 42 days or less, preferably 35 days or less, and more preferably 28 days or less. In the culture of the present invention, passages may be performed as many times as necessary to obtain the desired amount of regulatory T cells, and medium addition and replacement may be performed. The culture of the present invention can be performed using a known CO2 incubator. The culture vessel is not particularly limited and can be appropriately selected from plates, dishes, petri dishes, flasks, bags, bottles, tanks (culture vessels), bioreactors, etc. A vessel to which an anti-CD3 antibody is bound can be used as the culture vessel.

 工程(1)では、IL-2、IL-4、IL-33、TNFR2アゴニスト(更にIL-3、TGF-βRアゴニストを加えてもよい)の存在下で、CD30アゴニスト、CD3アゴニスト及びCD28アゴニストからなる群から選択される少なくとも1種の存在下で培養(細胞を刺激)を行ってもよい。より好ましくは、IL-2、IL-4、IL-33、TNFR2アゴニスト(更にIL-3、TGF-βRアゴニストを加えてもよい)の存在下で、最初はCD30アゴニスト、CD3アゴニスト及びCD28アゴニストからなる群から選択される少なくとも1種の存在下で培養(細胞を刺激)を開始した後、これらの抗体の非存在下で培養を行ってもよい。工程(1)の工程を(継代培養により)繰り返し行ってもよく、最初はCD30アゴニスト、CD3アゴニスト及びCD28アゴニストからなる群から選択される少なくとも1種の存在下で培養(細胞を刺激)を開始した後、これらの抗体の非存在下で培養を行うことを繰り返すようにして培養を行ってもよい。この繰り返しの培養期間は、例えば、7日~28日、好ましくは14日~21日である。具体的には、最初の3日程度をCD30アゴニスト、CD3アゴニスト及びCD28アゴニストからなる群から選択される少なくとも1種の存在下で細胞を培養(刺激)した後、これらを含まない培地で4~11日間程度培養を行い、この単位の培養を繰り返すことが挙げられる。培養の繰り返しの回数は、特に制限されず、当業者であれば本発明の免疫細胞の数などをモニターしながら適宜決定することが可能であり、例えば、1回以上、2回以上、3回以上、4回以上、5回以上、6回以上、7回以上である。培養の繰り返しの回数の上限は、例えば20回である。 In step (1), culture (cell stimulation) may be performed in the presence of IL-2, IL-4, IL-33, and a TNFR2 agonist (IL-3 and a TGF-βR agonist may also be added), and in the presence of at least one selected from the group consisting of a CD30 agonist, a CD3 agonist, and a CD28 agonist. More preferably, culture (cell stimulation) may be initially initiated in the presence of IL-2, IL-4, IL-33, and a TNFR2 agonist (IL-3 and a TGF-βR agonist may also be added), and in the presence of at least one selected from the group consisting of a CD30 agonist, a CD3 agonist, and a CD28 agonist, and then culture may be performed in the absence of these antibodies. Step (1) may be repeated (by subculture). The culture may be performed by first initiating culture (stimulating the cells) in the presence of at least one selected from the group consisting of a CD30 agonist, a CD3 agonist, and a CD28 agonist, followed by repeated culture in the absence of these antibodies. The culture period for this repetition is, for example, 7 to 28 days, preferably 14 to 21 days. Specifically, the cells may be cultured (stimulated) in the presence of at least one selected from the group consisting of a CD30 agonist, a CD3 agonist, and a CD28 agonist for the first 3 days or so, followed by culture in a medium that does not contain these agonists for 4 to 11 days, and this culture unit may be repeated. The number of culture repetitions is not particularly limited and can be determined appropriately by a person skilled in the art while monitoring the number of immune cells of the present invention, for example, 1 or more, 2 or more, 3 or more, 4 or more, 5 or more, 6 or more, or 7 or more. The upper limit of the number of culture repetitions is, for example, 20 times.

 CD30アゴニストとしては、CD30に特異的に結合することにより、CD30から細胞内にシグナルを伝達することができる分子であれば特に限定されない。CD30アゴニストとしては、例えば、抗CD30アゴニス卜抗体又はその機能性断片、CD30リガンド又はその機能性断片等が挙げられる。培地中におけるCD30アゴニストの濃度は、特に限定されず、用いるCD30アゴニストの種類などによって適宜調整される。CD30アゴニストの濃度は、例えば、10~1000ng/mL、好ましくは100~500ng/mLである。 The CD30 agonist is not particularly limited as long as it is a molecule that can transmit a signal from CD30 into the cell by specifically binding to CD30. Examples of CD30 agonists include anti-CD30 agonist antibodies or functional fragments thereof, and CD30 ligands or functional fragments thereof. The concentration of the CD30 agonist in the culture medium is not particularly limited and is adjusted appropriately depending on the type of CD30 agonist used. The concentration of the CD30 agonist is, for example, 10 to 1000 ng/mL, preferably 100 to 500 ng/mL.

 CD3アゴニストとしては、CD3に特異的に結合することにより、CD3から細胞内にシグナルを伝達することができる分子であれば特に限定されない。CD3アゴニストとしては、例えば、抗CD3アゴニス卜抗体又はその機能性断片、CD3リガンド又はその機能性断片等が挙げられる。培地中におけるCD3アゴニストの濃度は、特に限定されず、用いるCD3アゴニストの種類などによって適宜調整される。CD3アゴニストの濃度は、例えば、10~10000ng/mL、好ましくは100~10000ng/mL、より好ましくは1000~5000ng/mLである。 The CD3 agonist is not particularly limited, as long as it is a molecule that can transmit a signal from CD3 into the cell by specifically binding to CD3. Examples of CD3 agonists include anti-CD3 agonist antibodies or functional fragments thereof, and CD3 ligands or functional fragments thereof. The concentration of the CD3 agonist in the culture medium is not particularly limited and is adjusted appropriately depending on the type of CD3 agonist used. The concentration of the CD3 agonist is, for example, 10 to 10,000 ng/mL, preferably 100 to 10,000 ng/mL, and more preferably 1,000 to 5,000 ng/mL.

 CD28アゴニストとしては、CD28に特異的に結合することにより、CD28から細胞内にシグナルを伝達することができる分子であれば特に限定されない。CD28アゴニストとしては、例えば、抗CD28アゴニス卜抗体又はその機能性断片、CD28リガンド又はその機能性断片等が挙げられる。培地中におけるCD28アゴニストの濃度は、特に限定されず、用いるCD28アゴニストの種類などによって適宜調整される。CD28アゴニストの濃度は、例えば、10~10000ng/mL、好ましくは100~10000ng/mL、より好ましくは500~5000ng/mLである。 The CD28 agonist is not particularly limited, as long as it is a molecule that can transmit a signal from CD28 into the cell by specifically binding to CD28. Examples of CD28 agonists include anti-CD28 agonist antibodies or functional fragments thereof, and CD28 ligands or functional fragments thereof. The concentration of the CD28 agonist in the culture medium is not particularly limited and is adjusted appropriately depending on the type of CD28 agonist used. The concentration of the CD28 agonist is, for example, 10 to 10,000 ng/mL, preferably 100 to 10,000 ng/mL, and more preferably 500 to 5,000 ng/mL.

 本発明の製造方法で使用する制御性T細胞を含む細胞集団は、骨髄、臍帯血、血液などの生体組織から単離された初代培養細胞である。初代培養細胞とは、生体から分離した器官、細胞及び組織を播種し、第1回目の継代を行うまでの細胞を意味する。初代培養細胞は、臓器及び組織から酵素処理、物理的方法による分散、explant法等の公知の方法により得ることができる。初代制御性T細胞を含む細胞集団に含まれる制御性T細胞の割合(細胞数)は、例えば、80%以上、好ましくは90%以上、より好ましくは95%以上であり、また上限は、例えば、100%以下である。生体組織から分離された細胞からの制御性T細胞の単離は、フローサイトメトリーを用いた方法、磁気細胞分離法などの公知の方法により行うことができる。 The cell population containing regulatory T cells used in the production method of the present invention is primary cultured cells isolated from biological tissues such as bone marrow, umbilical cord blood, and blood. Primary cultured cells refer to cells that have been isolated from organs, cells, and tissues separated from a living body and seeded until the first passage. Primary cultured cells can be obtained from organs and tissues by known methods such as enzyme treatment, physical dispersion, and the explant method. The proportion (cell number) of regulatory T cells contained in the cell population containing primary regulatory T cells is, for example, 80% or more, preferably 90% or more, more preferably 95% or more, with an upper limit of, for example, 100% or less. Regulatory T cells can be isolated from cells separated from biological tissues by known methods such as flow cytometry or magnetic cell separation.

 本発明の製造方法で使用する、初代制御性T細胞を含む細胞集団は、ヒト由来であってもよいし、ヒト以外の哺乳類(非ヒト哺乳類)由来の細胞であってもよく、好ましくはヒトである。非ヒト哺乳類としては、例えば、マウス、ラット、ハムスター、モルモット、ウサギ、イヌ、ネコ、ブタ、ウシ、ウマ、ヒツジ、サルが挙げられる。 The cell population containing primary regulatory T cells used in the production method of the present invention may be derived from humans or from mammals other than humans (non-human mammals), preferably humans. Examples of non-human mammals include mice, rats, hamsters, guinea pigs, rabbits, dogs, cats, pigs, cows, horses, sheep, and monkeys.

 本発明の製造方法は、得られた制御性T細胞を濃縮するために、制御性T細胞を分離する工程を更に含んでいてもよい。制御性T細胞の分離は、フローサイトメトリーを用いた方法、磁気細胞分離法などの公知の方法により行うことができる。 The production method of the present invention may further include a step of separating regulatory T cells in order to enrich the obtained regulatory T cells. Separation of regulatory T cells can be carried out by known methods such as flow cytometry or magnetic cell separation.

 本発明の製造方法により、制御性T細胞が増殖した、制御性T細胞を含む細胞集団を製造することができる。すなわち、本発明の製造方法により制御性T細胞を拡大培養することができる。 The manufacturing method of the present invention makes it possible to produce a cell population containing regulatory T cells in which regulatory T cells have proliferated. In other words, the manufacturing method of the present invention makes it possible to expand and culture regulatory T cells.

 また、本発明の別の実施形態である、制御性T細胞を含む細胞集団の増殖方法(本明細書において、単に「本発明の増殖方法」と称することがある)は、以下の工程を含むことを特徴とする。
(1A)IL-2、IL―4、IL-33及びTNFR2アゴニストの存在下で初代制御性T細胞を含む細胞集団を培養する工程。
Furthermore, another embodiment of the present invention, a method for expanding a cell population containing regulatory T cells (sometimes referred to herein simply as the "expansion method of the present invention"), is characterized by comprising the following steps:
(1A) Culturing a cell population containing primary regulatory T cells in the presence of IL-2, IL-4, IL-33 and a TNFR2 agonist.

 工程(1A)は、工程(1)と同様に実施することができる。上記の制御性T細胞を含む細胞集団の増殖方法には、本発明の製造方法に関する説明が適用される。 Step (1A) can be carried out in the same manner as step (1). The explanation regarding the production method of the present invention applies to the method for expanding the above-mentioned cell population containing regulatory T cells.

 本発明の製造方法及び増殖方法により得られた制御性T細胞は、外来遺伝子が導入された制御性T細胞であってもよい。 The regulatory T cells obtained by the production method and proliferation method of the present invention may be regulatory T cells into which an exogenous gene has been introduced.

 「外来遺伝子」は、制御性T細胞に所望のタンパク質を発現させるために、外部より導入される遺伝子であり、制御性T細胞の用途に応じて適宜選択することができる。 An "exogenous gene" is a gene introduced from outside to cause regulatory T cells to express a desired protein, and can be selected appropriately depending on the intended use of the regulatory T cells.

 外来遺伝子は、例えば、CAR(キメラ抗原受容体)を発現するための遺伝子とすることができ、そこに更に、サイトカイン及び/又はケモカインを発現するための遺伝子を含むことができる。制御性T細胞が発現するCARは基本的に、一般的又は公知のCARと同様に、(i)がん細胞の細胞表面抗原を認識する抗原認識部位(例えば、一本鎖抗体)、(ii)細胞膜貫通領域、及び(iii)T細胞の活性化を誘導するシグナル伝達領域、の各部位のペプチドが、必要に応じてスペーサーを介して連結することによって構成されている。また、外来遺伝子は、例えば、外因性T細胞受容体(TCR)を発現するための遺伝子とすることもできる。外因性TCRとは、外因性TCRをコードする核酸が導入されるT細胞にとって外因性であることを意味し、外因性TCRのアミノ酸配列は当該T細胞の内因性TCRと同一であってもよく、異なっていてもよい。外来遺伝子は、1種類、又は2種類以上を導入することができる(例えば、CARと外因性TCR)。 The foreign gene can be, for example, a gene for expressing a chimeric antigen receptor (CAR), which can further include genes for expressing cytokines and/or chemokines. Similar to common or known CARs, the CAR expressed by regulatory T cells is essentially composed of peptides from each of the following sites, linked, as necessary, via a spacer: (i) an antigen recognition site (e.g., a single-chain antibody) that recognizes a cell surface antigen on a cancer cell; (ii) a cell membrane-spanning domain; and (iii) a signal transduction domain that induces T cell activation. The foreign gene can also be, for example, a gene for expressing an exogenous T cell receptor (TCR). An exogenous TCR means that the nucleic acid encoding the exogenous TCR is exogenous to the T cell into which it is introduced, and the amino acid sequence of the exogenous TCR may be the same as or different from the endogenous TCR of the T cell. One or more types of foreign genes can be introduced (e.g., a CAR and an exogenous TCR).

 外来遺伝子を細胞に導入するための手段は特に限定されるものではなく、公知又は一般的な各種の手段を採用することができる。典型的には外来遺伝子は、発現ベクターを用いて細胞に導入し、発現させる。発現ベクターは、直鎖状でも環状でもよく、プラスミドなどの非ウイルスベクターでも、ウイルスベクターでも、トランスポゾンによるベクターでもよい。なお、外来遺伝子を導入する細胞は、特に限定されず、いずれの段階であってもよく、例えば、工程(1)又は(1A)を実施する前の初代制御性T細胞、本発明の製造方法及び増殖方法により得られた制御性T細胞などが挙げられる。 The means for introducing a foreign gene into cells is not particularly limited, and various known or common means can be used. Typically, the foreign gene is introduced into cells using an expression vector and expressed. The expression vector may be linear or circular, and may be a non-viral vector such as a plasmid, a viral vector, or a transposon-based vector. The cells into which the foreign gene is introduced are not particularly limited, and may be at any stage. Examples include primary regulatory T cells before carrying out step (1) or (1A), and regulatory T cells obtained by the production method and expansion method of the present invention.

 発現ベクターを細胞に導入するための手法は、実施形態に応じた適切なものとすることができる。例えば、ウイルス感染法、カルシウムリン酸法、リポフェクション法、マイクロインジェクション法、エレクトロポレーション法などの公知の方法により、発現ベクターを細胞に導入することができる。発現ベクターは、公知の手段によって、また適宜市販のキットを用いて(その指示書に従って)、各手法における使用に適した形態に調製することができる。 The technique for introducing an expression vector into cells can be an appropriate one depending on the embodiment. For example, the expression vector can be introduced into cells by known methods such as viral infection, calcium phosphate precipitation, lipofection, microinjection, and electroporation. The expression vector can be prepared in a form suitable for use in each technique by known means or, as appropriate, using a commercially available kit (following the instructions).

 発現ベクターは、ウイルス感染法により細胞に導入することができる。ウイルスベクターとしては、例えば、レトロウイルスベクター、レンチウイルスベクター、アデノウイルスベクター、アデノ随伴ウイルスベクターが挙げられる。これらのウイルスベクターを用いる場合は、対応する市販のキットを用いて、上記外来遺伝子を含むベクター及び各ウイルスのパッケージングベクター(プラスミド)を宿主細胞にトランスフェクションして組換えウイルスを作製した後、得られた組換えウイルスを細胞に感染させるようにすればよい。 The expression vector can be introduced into cells by viral infection. Examples of viral vectors include retroviral vectors, lentiviral vectors, adenoviral vectors, and adeno-associated viral vectors. When using these viral vectors, a corresponding commercially available kit can be used to transfect host cells with the vector containing the foreign gene and the packaging vector (plasmid) of each virus to produce a recombinant virus, and then the resulting recombinant virus can be used to infect cells.

 発現ベクターは、上記外来遺伝子に加えて、必要に応じて、核局在化シグナル(NLS)、マルチクローニングサイト(MCS)等の配列を含んでいてもよい。発現ベクターは更に、レポーター遺伝子(例えば、各色の蛍光タンパク質をコードする遺伝子)、薬剤選択遺伝子(例えば、カナマイシン耐性遺伝子、アンピシリン耐性遺伝子、ピューロマイシン耐性遺伝子)、自殺遺伝子(例えば、ジフテリアA毒素、単純ヘルペスチミジンキナーゼ(HSV-TK)、カルボキシペプチダーゼG2(CPG2)、カルボキシルエステラーゼ(CA)、シトシンデアミナーゼ(CD)、チトクロームP450(cyt-450)、デオキシシチジンキナーゼ(dCK)、ニトロレダクターゼ(NR)、プリンヌクレオシドホスホリラーゼ(PNP)、チミジンホスホリラーゼ(TP)、水痘帯状疱疹ウイルスチミジンキナーゼ(VZV-TK)、キサンチン-グアニンホスホリボシルトランスフェラーゼ(XGPRT)、誘導性カスパーゼ9(inducible caspase 9)などをコードする遺伝子)のような“機能的遺伝子”をコードする核酸(塩基配列)を含んでいてもよい。 In addition to the foreign gene, the expression vector may contain sequences such as a nuclear localization signal (NLS) or a multiple cloning site (MCS) as necessary. The expression vector may further contain a reporter gene (e.g., a gene encoding a fluorescent protein of each color), a drug selection gene (e.g., a kanamycin resistance gene, an ampicillin resistance gene, a puromycin resistance gene), a suicide gene (e.g., a diphtheria A toxin, a herpes simplex thymidine kinase (HSV-TK), a carboxypeptidase G2 (CPG2), a carboxylesterase (CA), a cytosine deaminase (CD), a cytochrome P450 (cyt-450), It may also contain nucleic acids (base sequences) encoding "functional genes" such as genes encoding deoxycytidine kinase (dCK), nitroreductase (NR), purine nucleoside phosphorylase (PNP), thymidine phosphorylase (TP), varicella-zoster virus thymidine kinase (VZV-TK), xanthine-guanine phosphoribosyltransferase (XGPRT), inducible caspase 9, etc.

 本発明の製造方法及び増殖方法により製造した制御性T細胞を含む細胞集団は、免疫反応が異常に亢進している動物(特に、ヒト)の治療に有用であり、例えば、X染色体連鎖型免疫調節異常・多発性内分泌障害・腸症(IPEX)症候群、移植片対宿主病(GVHD)、臓器移植における拒絶反応、自己免疫疾患、炎症性疾患、アレルギー疾患(花粉症、喘息、アトピー性皮膚炎、湿疹、食品アレルギー、食物過敏症、蕁麻疹、アレルギー性鼻炎、アレルギー性結膜炎、薬剤アレルギー)などの治療及び予防に有用であるが、これらに限定されない。本発明の製造方法及び増殖方法により製造した制御性T細胞は、自家移植に用いてもよく、他家移植に用いてもよい。また、他の薬剤と併用してもよい。 A cell population containing regulatory T cells produced by the production method and expansion method of the present invention is useful for treating animals (particularly humans) with abnormally enhanced immune responses, including, but not limited to, X-linked immunodysregulation/polyendocrinopathy/enteropathy (IPEX) syndrome, graft-versus-host disease (GVHD), organ transplant rejection, autoimmune diseases, inflammatory diseases, and allergic diseases (hay fever, asthma, atopic dermatitis, eczema, food allergies, food hypersensitivity, urticaria, allergic rhinitis, allergic conjunctivitis, and drug allergies). Regulatory T cells produced by the production method and expansion method of the present invention may be used for autologous or allogeneic transplantation. They may also be used in combination with other drugs.

 このような細胞治療を実施する場合、拒絶反応が起こらないという観点から、制御性T細胞の製造に使用するための細胞が単離される対象は、制御性T細胞が投与される対象とHLAの型が一致していることが好ましく、制御性T細胞が投与される対象と同一の対象であることがより好ましい。 When carrying out such cell therapy, in order to avoid rejection reactions, it is preferable that the subject from whom the cells to be used in producing regulatory T cells are isolated has an HLA type that matches that of the subject to which the regulatory T cells will be administered, and it is even more preferable that the subject is the same as the subject to which the regulatory T cells will be administered.

 本発明によれば、制御性T細胞を含む細胞集団を含有してなる医薬(以下、本発明の医薬と称することがある)を製造することができる。本発明の医薬は、公知の手段(例えば、日本薬局方に記載の方法等)に従って、有効量の制御性T細胞を薬学的に許容される担体と混合するなどして、非経口製剤として製造することが好ましい。本発明の医薬は、注射剤、懸濁剤、点滴剤等の非経口製剤として製造することが好ましい。非経口的な投与方法としては、静脈内、動脈内、筋肉内、腹腔内、皮下投与などの方法が挙げられる。薬学的に許容される担体としては、例えば、溶剤、基剤、希釈剤、賦形剤、無痛化剤、緩衝剤、保存料、安定剤、懸濁剤、等張化剤、界面活性剤、溶解補助剤等が挙げられる。 According to the present invention, it is possible to produce a pharmaceutical comprising a cell population that includes regulatory T cells (hereinafter, sometimes referred to as the pharmaceutical of the present invention). The pharmaceutical of the present invention is preferably produced as a parenteral formulation by mixing an effective amount of regulatory T cells with a pharmaceutically acceptable carrier according to known means (for example, the method described in the Japanese Pharmacopoeia). The pharmaceutical of the present invention is preferably produced as a parenteral formulation such as an injection, suspension, or infusion. Parenteral administration methods include intravenous, intraarterial, intramuscular, intraperitoneal, and subcutaneous administration. Pharmaceutically acceptable carriers include, for example, solvents, bases, diluents, excipients, soothing agents, buffers, preservatives, stabilizers, suspending agents, isotonicity agents, surfactants, and solubilizing agents.

 本発明の医薬の投与量は、患者の体重、年齢、性別、症状などの種々の条件に応じて適宜決定することができるが、通常、細胞数として、体重60kgの対象に対し、1回当り、通常1×10~1×1010個となるように、好ましくは1×10~1×10個となるように、より好ましくは5×10~5×10個となるように投与される。また、1回で投与してもよく、複数回にわたって投与してもよい。本発明の医薬は、非経口投与に適した公知の形態、例えば、注射又は注入剤とすることができる。また、本発明の医薬は、細胞を安定に維持するために、生理食塩水、リン酸緩衝生理食塩水(PBS)、培地等を含んでもよい。培地としては、特に限定するものではないが、RPMI、AIM-V、X-VIVO10などの培地が挙げられるが、これらに限定されない。また、該医薬には医薬的に許容される担体(例:ヒト血清アルブミン)、保存剤等が安定化の目的で添加されていてもよい。本発明の医薬は、ヒトを含む哺乳動物に対して適用されるものである。 The dosage of the medicament of the present invention can be determined appropriately depending on various conditions, such as the patient's body weight, age, sex, and symptoms. Typically, the number of cells administered per administration to a subject weighing 60 kg is typically 1×10 6 to 1×10 10 , preferably 1×10 7 to 1×10 9 , and more preferably 5×10 7 to 5×10 8. The medicament may be administered once or multiple times. The medicament of the present invention can be in a known form suitable for parenteral administration, such as an injection or infusion. Furthermore, the medicament of the present invention may contain physiological saline, phosphate-buffered saline (PBS), a culture medium, or the like, to stably maintain the cells. Examples of culture media include, but are not limited to, RPMI, AIM-V, and X-VIVO10. Furthermore, the medicament may contain a pharmaceutically acceptable carrier (e.g., human serum albumin), a preservative, or the like, for stabilization purposes. The medicament of the present invention is intended for use in mammals, including humans.

 本明細書及び請求の範囲で使用される場合、特に文脈上必要とされない限り、単数形の用語は複数形を含み、複数形の用語は単数形を含むものとする。したがって、単数形の冠詞(例えば、英語の場合は「a」、「an」、「the」など)は、特に言及しない限り、その複数形の概念をも含むことが理解されるべきである。 As used in this specification and claims, singular terms shall include plurals and plural terms shall include the singular, unless the context otherwise requires. Therefore, singular articles (e.g., "a," "an," "the," etc. in English) should be understood to include the plural concept, unless otherwise specified.

 以下、本発明を更に詳しく説明するため実施例を挙げる。しかし、本発明はこれら実施例等になんら限定されるものではない。 The following examples are provided to further explain the present invention. However, the present invention is not limited to these examples.

 (1)初代培養Tregの単離
 ヒト末梢血単核球分画(PBMC(HemaCare))より、CD4T細胞をCD4T cell isolation kit Human(Myltenyi Biotec)の推奨プロトコルに従って単離した後、以下の抗体セットを用いて染色し、フローサイトメトリーによって解析とTreg(CD4CD8αCD25CD127)の精製を行った。結果を図1に示す。
PE/Cy7 CD4、PerCP/Cy5.5 CD8α、PE CD127、APC CD25。
(1) Isolation of primary cultured Tregs CD4 + T cells were isolated from human peripheral blood mononuclear cells (PBMCs (HemaCare)) according to the recommended protocol of the CD4 + T cell isolation kit Human (Myltenyi Biotec). Then, they were stained with the following antibody set and analyzed by flow cytometry, and Tregs (CD4 + CD8α - CD25 + CD127 - ) were purified. The results are shown in Figure 1.
PE/Cy7 CD4, PerCP/Cy5.5 CD8α, PE CD127, APC CD25.

 精製後のTregは99.0%の精製度で得られた。 Purified Tregs were obtained with a purity of 99.0%.

 (2)初代培養Tregの拡大培養
 (1)で得られた初代培養Tregを、抗CD3抗体(3μg/mL,eBioscience)を結合させた96穴細胞培養プレートに1×10cells/wellで播種し、COインキュベーターにて37℃、5.0%CO条件下で3日間培養した後、48穴細胞培養プレートに播き直して培養を続けた。さらに3日間培養した後、24穴G-Rex細胞培養プレートに播き直して培養を続けた。培地には終濃度でFBS(15%,Corning)、L-グルタミン-ペニシリン-ストレプトマイシン溶液(1/100,Invitrogen,Sigma-Aldrich)、インスリン-トランスフェリン-セレン サプリメント(1/100,Invitrogen)、アスコルビン酸2-リン酸(50μg/mL,Sigma-Aldrich)、IL-2(100ng/mL,PeproTech)、抗TNFR2抗体(3μg/mL,Hycult Biotech)、IL-3(60ng/mL,BioLegend)、IL-4(30ng/mL,BioLegend)、IL-33(30ng/mL,BioLegend)、及びTGF-β1(5ng/mL,BioLegend)を含んだα-MEM(Invitrogen)を用いた。最初の3日間は抗CD28抗体(1.5μg/mL,BioLegend)、抗CD30抗体(300ng/mL,R&D Systems)を上記の組成に対してさらに添加した。また、コントロール群として、上記培地組成から抗TNFR2抗体、IL-3、IL-4、IL-33及びTGF-β1を抜いた培地を用いた。培養は培養開始3日目、6日目、8日目、11日目、13日目に培地交換しながら15日間培養し、細胞数を計測して折れ線グラフを作成した。結果を図2に示す。
(2) Expansion of Primary Cultured Tregs The primary cultured Tregs obtained in (1) were seeded at 1 x 10 cells/well onto a 96-well cell culture plate conjugated with anti-CD3 antibody (3 μg/mL, eBioscience) and cultured for 3 days at 37°C and 5.0% CO in a CO incubator. After further culture for 3 days, the cells were seeded onto a 48-well cell culture plate and continued to be cultured. After further culture for 3 days, the cells were seeded onto a 24-well G-Rex cell culture plate and continued to be cultured. The medium contained, at final concentrations, FBS (15%, Corning), L-glutamine-penicillin-streptomycin solution (1/100, Invitrogen, Sigma-Aldrich), insulin-transferrin-selenium supplement (1/100, Invitrogen), ascorbic acid 2-phosphate (50 μg/mL, Sigma-Aldrich), IL-2 (100 ng/mL, PeproTech), and anti-TNFR2 antibody (3 μg/mL, Hycult The α-MEM (Invitrogen) medium used contained IL-1 (60 ng/mL, BioLegend), IL-3 (60 ng/mL, BioLegend), IL-4 (30 ng/mL, BioLegend), IL-33 (30 ng/mL, BioLegend), and TGF-β1 (5 ng/mL, BioLegend). For the first three days, anti-CD28 antibody (1.5 μg/mL, BioLegend) and anti-CD30 antibody (300 ng/mL, R&D Systems) were further added to the above composition. A control group used medium containing the above medium composition but omitting anti-TNFR2 antibody, IL-3, IL-4, IL-33, and TGF-β1. The cells were cultured for 15 days, with the medium being changed on days 3, 6, 8, 11, and 13 after the start of culture, and the number of cells was counted and a line graph was created. The results are shown in Figure 2.

 拡大培養を経て得られた細胞集団は培養14日目に約5,700倍の増殖力を示した。また、コントロール群と比較して増殖が亢進している結果となった。 The cell population obtained through expansion culture showed a proliferation rate of approximately 5,700 times on the 14th day of culture. Furthermore, proliferation was enhanced compared to the control group.

 (3)拡大培養におけるTregのタンパク質の発現量の検討
 (2)でIL-2、IL-3、IL―4、IL-33、TGF-β1及び抗TNFR2抗体を含む培地で拡大培養をした、培養開始後15日目の各細胞集団を用いて、以下の抗体セットを用いて染色し、フローサイトメトリーによって解析した。結果を図3に示す。Zombie NIR,BV510 CD3,BV421 CD4、PE/Cy7 CD8β,APC CD25,FITC FOXP3,PE Helios。
(3) Examination of Treg protein expression levels during expansion culture. Each cell population, which was expanded in (2) in a medium containing IL-2, IL-3, IL-4, IL-33, TGF-β1, and anti-TNFR2 antibody, was used on day 15 after the start of culture. The cells were stained with the following antibody set and analyzed by flow cytometry. The results are shown in Figure 3: Zombie NIR, BV510 CD3, BV421 CD4, PE/Cy7 CD8β, APC CD25, FITC FOXP3, PE Helios.

 上記の方法で得られた細胞集団は77.6%のFOXP3陽性率が見られた。また、Heliosの陽性率も59.5%を示した。 The cell population obtained using the above method showed a FOXP3 positivity rate of 77.6%. The Helios positivity rate was also 59.5%.

 (4)Tregの抑制性機能の評価
 Tregとは別ドナー由来の抗CD3抗体未処置又は処置したPBMCをCellTrace Violet(Thermo Fisher Scientific)で染色したものをターゲット細胞として用い、(2)でIL-2、IL-3、IL―4、IL-33、TGF-β1及び抗TNFR2抗体を含む培地で拡大培養をしたTregと共培養した。培地には終濃度でFBS(15%,Corning)、L-グルタミン-ペニシリン-ストレプトマイシン溶液(1/100,Invitrogen,Sigma-Aldrich)、インスリン-トランスフェリン-セレン サプリメント(1/100,Invitrogen)、及びアスコルビン酸2-リン酸(50μg/mL,Sigma-Aldrich)を含んだα-MEM(Invitrogen)を用い、4日間培養した。各細胞集団は以下の抗体セットを用いて染色した。結果を図4に示す。
Zombie NIR,PE/Cy7 CD3,FITC HLA-A24。
(4) Evaluation of the suppressive function of Tregs. PBMCs derived from a donor other than Tregs and untreated or treated with anti-CD3 antibody were stained with CellTrace Violet (Thermo Fisher Scientific) and used as target cells. These cells were co-cultured with Tregs expanded in a medium containing IL-2, IL-3, IL-4, IL-33, TGF-β1, and anti-TNFR2 antibody in (2). The cells were cultured for 4 days in α-MEM (Invitrogen) containing final concentrations of FBS (15%, Corning), L-glutamine-penicillin-streptomycin solution (1/100, Invitrogen, Sigma-Aldrich), insulin-transferrin-selenium supplement (1/100, Invitrogen), and ascorbic acid 2-phosphate (50 μg/mL, Sigma-Aldrich). Each cell population was stained with the following antibody set. The results are shown in Figure 4.
Zombie NIR, PE/Cy7 CD3, FITC HLA-A24.

 Tregの細胞数依存的にターゲット細胞(CD3)の細胞分裂の強い抑制が見られた。 A strong suppression of target cell (CD3 + ) cell division was observed in a Treg cell number-dependent manner.

 (5)初代培養Tregの拡大培養
 (1)で得られた初代培養Tregを、抗CD3抗体(3μg/mL,eBioscience)を結合させた96穴細胞培養プレートに1×10cells/wellで播種し、COインキュベーターにて37℃、5.0%CO条件下で3日間培養した後、48穴細胞培養プレートに播き直して培養を続けた。さらに3日間培養した後、24穴G-Rex細胞培養プレートに播き直して培養を続けた。培地には終濃度でFBS(15%,Corning)、L-グルタミン-ペニシリン-ストレプトマイシン溶液(1/100,Invitrogen,Sigma-Aldrich)、インスリン-トランスフェリン-セレン サプリメント(1/100,Invitrogen)、アスコルビン酸2-リン酸(50μg/mL,Sigma-Aldrich)IL-2(100ng/mL,PeproTech)、抗TNFR2抗体(3μg/mL,Hycult Biotech)、IL-3(60ng/mL,BioLegend)、IL-4(30ng/mL、BioLegend)、IL-33(30ng/mL,BioLegend)、及びTGF-β1(5ng/mL、BioLegend)を含んだα-MEM(Invitrogen)を用いた。最初の3日間は抗CD28抗体(1.5μg/mL,BioLegend)、抗CD30抗体(300ng/mL,R&D Systems)を上記の組成に対してさらに添加した。ただし、実際には、上記培地組成中、IL-2を含むが、IL-3、IL-4、IL-33、TGF-β1及び抗TNFR2抗体を含まない培地(IL-2のみを含む培地)、或いは上記培地組成からIL-3、IL-4、IL-33及びTGF-β1のいずれか1つを抜いた培地を用いた。培養は培養開始3日目、6日目、8日目、11日目、13日目に培地交換しながら14日間培養し、細胞数を計測して折れ線グラフを作成した。結果を図5に示す。
(5) Expansion of Primary Cultured Tregs The primary cultured Tregs obtained in (1) were seeded at 1 x 10 cells/well onto a 96-well cell culture plate conjugated with anti-CD3 antibody (3 μg/mL, eBioscience) and cultured for 3 days at 37°C and 5.0% CO in a CO incubator. After further culture for 3 days, the cells were seeded onto a 48-well cell culture plate and continued to be cultured. After further culture for 3 days, the cells were seeded onto a 24-well G-Rex cell culture plate and continued to be cultured. The medium contained, at final concentrations, FBS (15%, Corning), L-glutamine-penicillin-streptomycin solution (1/100, Invitrogen, Sigma-Aldrich), insulin-transferrin-selenium supplement (1/100, Invitrogen), ascorbic acid 2-phosphate (50 μg/mL, Sigma-Aldrich), IL-2 (100 ng/mL, PeproTech), and anti-TNFR2 antibody (3 μg/mL, Hycult). The culture medium used was α-MEM (Invitrogen) containing IL-3 (60 ng/mL, BioLegend), IL-4 (30 ng/mL, BioLegend), IL-33 (30 ng/mL, BioLegend), and TGF-β1 (5 ng/mL, BioLegend). For the first 3 days, anti-CD28 antibody (1.5 μg/mL, BioLegend) and anti-CD30 antibody (300 ng/mL, R&D Systems) were further added to the above composition. However, in practice, a medium containing IL-2 but not IL-3, IL-4, IL-33, TGF-β1, or anti-TNFR2 antibody (medium containing only IL-2) was used, or a medium obtained by removing any one of IL-3, IL-4, IL-33, and TGF-β1 from the medium composition. The culture was continued for 14 days, with medium changes on days 3, 6, 8, 11, and 13 after the start of culture, and the number of cells was counted and a line graph was created. The results are shown in FIG. 5.

 拡大培養を経て得られた細胞集団は培養14日目に、上記の培地条件からIL-4を抜いた条件、またIL-2を抜いた条件では増殖力が減少する結果となった。 The cell population obtained through expansion culture showed reduced proliferation ability on the 14th day of culture when IL-4 and IL-2 were removed from the above medium conditions.

 (6)拡大培養におけるTregのタンパク質の発現量の検討
 (5)で拡大培養をした、培養開始後14日目の各細胞集団(培養開始後14日目まで維持できなかったIL-2のみを含む培地で拡大培養した細胞収集団を除く)を用いて、以下の抗体セットを用いて染色し、フローサイトメトリーによって解析した。結果を図6に示す。Zombie NIR,BV510 CD3,BV421 CD4、PE/Cy7 CD8β,APC CD25,FITC FOXP3。
(6) Examination of Treg protein expression levels during expansion culture. Each cell population expanded in (5) on day 14 after the start of culture (excluding the cell collection expanded in medium containing only IL-2, which could not be maintained until day 14 after the start of culture) was stained with the following antibody set and analyzed by flow cytometry. The results are shown in Figure 6: Zombie NIR, BV510 CD3, BV421 CD4, PE/Cy7 CD8β, APC CD25, FITC FOXP3.

 (5)の培地組成からIL-3、IL-4、IL-33、及びTGF-β1のいずれかのサイトカインを抜いた培地で拡大培養をした各細胞集団中のCD25FOXP3集団はそれぞれ、62.8%、50.5%、65.8%、30.7%を示した。 When the CD25 + FOXP3 + population was expanded in a medium containing the cytokines IL-3, IL-4, IL-33, and TGF-β1 from the medium composition of (5), the percentages of the CD25 + FOXP3 + population in each cell population were 62.8%, 50.5%, 65.8%, and 30.7%, respectively.

 (7)Tregの抑制性機能の評価
 Tregとは別ドナー由来の抗CD3抗体未処置又は処置したPBMCをCellTrace Violet(Thermo Fisher Scientific)で染色したものをターゲット細胞として用い、(5)で培地組成からIL-3、IL-4、IL-33、及びTGF-β1のいずれかのサイトカインを抜いた培地で拡大培養をしたTregと共培養した。培地には終濃度でFBS(15%,Corning)、L-グルタミン-ペニシリン-ストレプトマイシン溶液(1/100,Invitrogen,Sigma-Aldrich)、インスリン-トランスフェリン-セレン サプリメント(1/100,Invitrogen)、及びアスコルビン酸2-リン酸(50μg/mL,Sigma-Aldrich)を含んだα-MEM(Invitrogen)を用い、4日間培養した。各細胞集団は以下の抗体セットを用いて染色した。結果を図7に示す。
Zombie NIR,PE/Cy7 CD3,FITC HLA-A24。
(7) Evaluation of the suppressive function of Tregs. PBMCs derived from a donor other than Tregs and untreated or treated with anti-CD3 antibodies were stained with CellTrace Violet (Thermo Fisher Scientific) and used as target cells. These cells were co-cultured with Tregs expanded in a medium containing no cytokines (IL-3, IL-4, IL-33, or TGF-β1) as described in (5). The cells were cultured for 4 days in α-MEM (Invitrogen) containing final concentrations of FBS (15%, Corning), L-glutamine-penicillin-streptomycin solution (1/100, Invitrogen, Sigma-Aldrich), insulin-transferrin-selenium supplement (1/100, Invitrogen), and ascorbic acid 2-phosphate (50 μg/mL, Sigma-Aldrich). Each cell population was stained with the following antibody set. The results are shown in Figure 7.
Zombie NIR, PE/Cy7 CD3, FITC HLA-A24.

 Tregの細胞数依存的にターゲット細胞(CD3)の細胞分裂の強い抑制が見られた。 A strong suppression of target cell (CD3 + ) cell division was observed in a Treg cell number-dependent manner.

 (8)初代培養Tregの拡大培養
 (1)で得られた初代培養Tregを、抗CD3抗体(3μg/mL,eBioscience)を結合させた96穴細胞培養プレートに1×10cells/wellで播種し、COインキュベーターにて37℃、5.0%CO条件下で3日間培養した後、48穴細胞培養プレートに播き直して培養を続けた。さらに3日間培養した後、24穴G-Rex細胞培養プレートに播き直して培養を続けた。培地には終濃度でFBS(15%,Corning)、L-グルタミン-ペニシリン-ストレプトマイシン溶液(1/100,Invitrogen,Sigma-Aldrich)、インスリン-トランスフェリン-セレン サプリメント(1/100,Invitrogen)、アスコルビン酸2-リン酸(50μg/mL,Sigma-Aldrich)、IL-2(100ng/mL,PeproTech)、抗TNFR2抗体(3μg/mL,Hycult Biotech)、IL-3(60ng/mL,BioLegend)、IL-4(30ng/mL、BioLegend)、IL-33(30ng/mL,BioLegend)、及びTGF-β1(5ng/mL、BioLegend)を含んだα-MEM(Invitrogen)を用いた。最初の3日間は抗CD28抗体(1.5μg/mL,BioLegend)、抗CD30抗体(300ng/mL,R&D Systems)を上記の組成に対してさらに添加した。また上記培地組成からIL-3、TGF-β1及び抗TNFR2抗体を抜いた培地も用いた。培養は培養開始3日目、6日目、8日目、11日目、13日目、15日目、18日目、20日目に培地交換しながら22日間培養し、細胞数を計測して折れ線グラフを作成した。結果を図8に示す。
(8) Expansion of Primary Cultured Tregs The primary cultured Tregs obtained in (1) were seeded at 1 x 10 cells/well onto a 96-well cell culture plate conjugated with anti-CD3 antibody (3 μg/mL, eBioscience) and cultured for 3 days at 37°C and 5.0% CO in a CO incubator. After further culture for 3 days, the cells were seeded onto a 48-well cell culture plate and continued to be cultured. After further culture for 3 days, the cells were seeded onto a 24-well G-Rex cell culture plate and continued to be cultured. The medium contained, at final concentrations, FBS (15%, Corning), L-glutamine-penicillin-streptomycin solution (1/100, Invitrogen, Sigma-Aldrich), insulin-transferrin-selenium supplement (1/100, Invitrogen), ascorbic acid 2-phosphate (50 μg/mL, Sigma-Aldrich), IL-2 (100 ng/mL, PeproTech), and anti-TNFR2 antibody (3 μg/mL, Hycult The culture medium used was α-MEM (Invitrogen) containing IL-3 (60 ng/mL, BioLegend), IL-4 (30 ng/mL, BioLegend), IL-33 (30 ng/mL, BioLegend), and TGF-β1 (5 ng/mL, BioLegend). For the first three days, anti-CD28 antibody (1.5 μg/mL, BioLegend) and anti-CD30 antibody (300 ng/mL, R&D Systems) were further added to the above composition. A medium containing the above medium composition but omitting IL-3, TGF-β1, and anti-TNFR2 antibody was also used. The culture was continued for 22 days, with the medium being changed on days 3, 6, 8, 11, 13, 15, 18, and 20 after the start of culture, and the number of cells was counted and a line graph was created. The results are shown in Figure 8.

 上記の培地条件からIL-3、TGF-β1及び抗TNFR2抗体を抜いた培地条件よりも、上記の培地条件の方が増殖力が強い結果となった。 The above medium conditions resulted in stronger proliferation than medium conditions in which IL-3, TGF-β1, and anti-TNFR2 antibody were omitted.

 本出願は、日本で2024年3月25日に出願された特願2024-048579号を基礎としており、その内容は本明細書にすべて包含される。 This application is based on Japanese Patent Application No. 2024-048579, filed on March 25, 2024, the contents of which are incorporated in their entirety herein.

Claims (16)

 制御性T細胞が増殖した細胞集団の製造方法であって、
(1)インターロイキン(IL)-2、IL-4、IL-33及びTNFR2アゴニストの存在下で初代制御性T細胞を含む細胞集団を培養する工程を含む方法。
A method for producing a cell population in which regulatory T cells are expanded, comprising:
(1) A method comprising the step of culturing a cell population containing primary regulatory T cells in the presence of interleukin (IL)-2, IL-4, IL-33 and a TNFR2 agonist.
 前記工程(1)を、
更にTGF-βRアゴニストの存在下で行う、請求項1に記載の方法。
The step (1)
The method according to claim 1, further carried out in the presence of a TGF-βR agonist.
 前記工程(1)を、
更にIL-3の存在下で行う、請求項1に記載の方法。
The step (1)
The method according to claim 1, further carried out in the presence of IL-3.
 前記工程(1)を、
更にIL-3及びTGF-βRアゴニストの存在下で行う、請求項1に記載の方法。
The step (1)
The method according to claim 1, further carried out in the presence of IL-3 and a TGF-βR agonist.
 前記TNFR2アゴニストが、抗TNFR2アゴニスト抗体である、請求項1に記載の方法。 The method of claim 1, wherein the TNFR2 agonist is an anti-TNFR2 agonist antibody.  前記TGF-βRアゴニストが、TGF-βである、請求項2又は4に記載の方法。 The method of claim 2 or 4, wherein the TGF-βR agonist is TGF-β.  前記工程(1)の培養において、更にCD30アゴニスト、CD3アゴニスト及びCD28アゴニストからなる群から選択される少なくとも1種の存在下で培養を行う、請求項1に記載の方法。 The method of claim 1, wherein the culture in step (1) is further performed in the presence of at least one agonist selected from the group consisting of a CD30 agonist, a CD3 agonist, and a CD28 agonist.  前記制御性T細胞が、CD25/FOXP3細胞である、請求項1に記載の方法。 The method of claim 1 , wherein the regulatory T cells are CD25 + /FOXP3 + cells.  前記CD25/FOXP3細胞が、CD4である、請求項8に記載の方法。 The method of claim 8, wherein the CD25 + /FOXP3 + cells are CD4 + .  制御性T細胞を含む細胞集団の増殖方法であって、
(1A)IL-2、IL-4、IL-33及びTNFR2アゴニストの存在下で初代制御性T細胞を含む細胞集団を培養する工程を含む方法。
A method for expanding a cell population containing regulatory T cells, comprising:
(1A) A method comprising the step of culturing a cell population containing primary regulatory T cells in the presence of IL-2, IL-4, IL-33 and a TNFR2 agonist.
 請求項1又は10に記載の方法により得られた、制御性T細胞を含む細胞集団。 A cell population containing regulatory T cells obtained by the method of claim 1 or 10.  請求項11に記載の制御性T細胞を含む細胞集団を含有してなる、医薬。 A pharmaceutical comprising a cell population containing the regulatory T cells described in claim 11.  免疫反応の異常な亢進の予防及び/又は治療に用いるための、請求項12に記載の医薬。 The pharmaceutical agent described in claim 12 for use in the prevention and/or treatment of abnormally enhanced immune responses.  請求項11に記載の制御性T細胞を含む細胞集団を、それを必要とする対象に投与することを含む、免疫反応の異常な亢進の予防及び/又は治療方法。 A method for preventing and/or treating abnormally enhanced immune responses, comprising administering a cell population containing the regulatory T cells described in claim 11 to a subject in need thereof.  免疫反応の異常な亢進の予防及び/又は治療において使用するための、請求項11に記載の制御性T細胞を含む細胞集団。 A cell population comprising the regulatory T cells of claim 11 for use in the prevention and/or treatment of abnormally enhanced immune responses.  免疫反応の異常な亢進の予防及び/又は治療するための医薬の製造における、請求項11に記載の制御性T細胞を含む細胞集団の使用。 Use of a cell population containing regulatory T cells described in claim 11 in the manufacture of a medicament for preventing and/or treating abnormally enhanced immune responses.
PCT/JP2025/011469 2024-03-25 2025-03-24 Method for propagating regulatory cells Pending WO2025205612A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2024-048579 2024-03-25
JP2024048579 2024-03-25

Publications (1)

Publication Number Publication Date
WO2025205612A1 true WO2025205612A1 (en) 2025-10-02

Family

ID=97215452

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2025/011469 Pending WO2025205612A1 (en) 2024-03-25 2025-03-24 Method for propagating regulatory cells

Country Status (1)

Country Link
WO (1) WO2025205612A1 (en)

Similar Documents

Publication Publication Date Title
US20250163132A1 (en) Compositions and methods for t cell delivery of therapeutic molecules
US20230110342A1 (en) Compositions and methods of stimulating and expanding t cells
JP2020518256A (en) Compositions and methods for gene editing in T cells using CRISPR/CPF1
CN112877291A (en) Genetically modified T cell products, methods of making and uses thereof
EP4410965A1 (en) Method for producing t cell
TW202345878A (en) Method for manufacturing regulatory t cell
EP4600352A1 (en) T cell production method
WO2025205612A1 (en) Method for propagating regulatory cells
WO2025205613A1 (en) Immune cells
HK1247957B (en) Compositions and methods for t cell delivery of therapeutic molecules
HK1243334B (en) Compositions and methods of stimulating and expanding t cells

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 25777500

Country of ref document: EP

Kind code of ref document: A1